Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters

Publication year range
1.
Int J Mol Sci ; 25(11)2024 May 27.
Article in English | MEDLINE | ID: mdl-38892029

ABSTRACT

Cardiometabolic diseases (CMDs) encompass a range of prevalent, often preventable, non-communicable illnesses, including myocardial infarction, stroke, cardiac insufficiency, arterial hypertension, obesity, type 2 diabetes mellitus, insulin resistance, chronic renal dysfunction, non-alcoholic fatty liver disease, and rare metabolic disorders [...].


Subject(s)
Cardiovascular Diseases , Humans , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/etiology , Metabolic Diseases/metabolism , Metabolic Diseases/etiology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/etiology , Animals , Insulin Resistance , Obesity/metabolism
2.
Rev Cardiovasc Med ; 24(7): 190, 2023 Jul.
Article in English | MEDLINE | ID: mdl-39077008

ABSTRACT

A variety of vegetable and fruit derived food oils are considered beneficial for human health due to their content of functional components including their positive effects in cardiovascular system. In addition to the favorable ratio of unsaturated versus saturated fatty acids, some of these oils include also other health beneficial compounds such as vitamins, minerals, pigments, enzymes and phenolic compounds. Particularly polyphenols have been documented to exert numerous positive effects in cardiovascular system including their anti-hypertensive, anti-atherogenic as well as cardio- and vasculo- protective effects in subjects suffering from various cardiovascular and cardiometabolic diseases, likely via their antioxidant, anti-inflammatory, anti-coagulant, anti-proliferative and anti-diabetic properties. However, it has not been proven so far whether the positive cardiovascular effects of polyphenol-rich food oils are, and to what measure, attributed to their phenolic content. Thus, the current review aims to summarize the main cardiovascular effects of major polyphenol-rich food oils including olive, flaxseed, soybean, sesame and coconut oils, and to uncover the role of their phenolic compounds in these effects.

3.
Int J Mol Sci ; 23(14)2022 Jul 13.
Article in English | MEDLINE | ID: mdl-35887089

ABSTRACT

Cardiovascular diseases (CVDs) are the top cause of death worldwide, and arterial hypertension per se remains the major preventable cause of CVDs [...].


Subject(s)
Cardiovascular Diseases , Hypertension , Cardiovascular Diseases/etiology , Cardiovascular Diseases/therapy , Humans , Hypertension/etiology
4.
Int J Mol Sci ; 23(24)2022 Dec 16.
Article in English | MEDLINE | ID: mdl-36555696

ABSTRACT

The aim of this study was to investigate the effects of quercetin (QUE) on the testicular architecture as well as markers of oxidative, inflammatory, and apoptotic profile of male gonads in Zucker diabetic fatty (ZDF) rats suffering from Type 2 diabetes mellitus in the absence or presence of obesity. QUE was administered orally at a dose of 20 mg/kg/day for 6 weeks. Morphometric analysis revealed that QUE treatment led to an improvement in testicular appearance, particularly in the case of Obese ZDF rats. Furthermore, a significant stabilization of the antioxidant capacity (p < 0.05), superoxide dismutase and catalase activity (p < 0.01), with a concomitant decrease in lipid peroxidation (p < 0.05) were observed in Obese ZDF animals exposed to QUE. Our data also indicate a significant decline in the levels of interleukin (IL)-1 (p < 0.05), IL-6 (p < 0.01) and tumor necrosis factor alpha (p < 0.001) following QUE supplementation to Obese ZDF rats in comparison with their respective control. Finally, a significant down-regulation of the pro-apoptotic BAX protein (p < 0.0001) was observed in Obese ZDF rats administered with QUE, while a significant Bcl-2 protein overexpression (p < 0.0001) was recorded in Lean ZDF animals when compared to their untreated control. As such, our results suggest that QUE is a potentially beneficial agent to reduce testicular damage in ZDF rats with Type 2 diabetes mellitus by decreasing oxidative stress, chronic inflammation, and excessive cell loss through apoptosis.


Subject(s)
Antioxidants , Diabetes Mellitus, Type 2 , Animals , Rats , Male , Antioxidants/pharmacology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Quercetin/pharmacology , Quercetin/therapeutic use , Rats, Zucker , Obesity/complications , Obesity/drug therapy , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use
5.
Int J Mol Sci ; 22(9)2021 Apr 24.
Article in English | MEDLINE | ID: mdl-33923282

ABSTRACT

Several mechanisms may contribute to cardiovascular pathology associated with diabetes, including dysregulation of matrix metalloproteinases (MMPs). Quercetin (QCT) is a substance with preventive effects in treatment of cardiovascular diseases and diabetes. The aim of the present study was to explore effects of chronic QCT administration on changes in heart function in aged lean and obese Zucker Diabetic Fatty (ZDF) rats and that in association with MMPs. Signaling underlying effects of diabetes and QCT were also investigated. In the study, we used one-year-old lean and obese ZDF rats treated for 6 weeks with QCT. Results showed that obesity worsened heart function and this was associated with MMP-2 upregulation, MMP-28 downregulation, and inhibition of superoxide dismutases (SODs). Treatment with QCT did not modulate diabetes-induced changes in heart function and MMPs. However, QCT activated Akt kinase and reversed effects of diabetes on SODs inhibition. In conclusion, worsened heart function due to obesity involved changes in MMP-2 and MMP-28 and attenuation of antioxidant defense by SOD. QCT did not have positive effects on improvement of heart function or modulation of MMPs. Nevertheless, its application mediated activation of adaptive responses against oxidative stress through Akt kinase and prevention of diabetes-induced negative effects on antioxidant defense by SODs.


Subject(s)
Cardiovascular Diseases/drug therapy , Diabetes Mellitus, Experimental/complications , Matrix Metalloproteinase 2/metabolism , Obesity/physiopathology , Oxidative Stress/drug effects , Quercetin/pharmacology , Aging , Animals , Antioxidants/pharmacology , Blood Glucose/metabolism , Cardiovascular Diseases/etiology , Cardiovascular Diseases/pathology , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinases, Secreted/genetics , Rats , Rats, Zucker
6.
Int J Mol Sci ; 22(17)2021 Sep 02.
Article in English | MEDLINE | ID: mdl-34502448

ABSTRACT

Cardioprotective medications are still unmet clinical needs. We have previously identified several cardioprotective microRNAs (termed ProtectomiRs), the mRNA targets of which may reveal new drug targets for cardioprotection. Here we aimed to identify key molecular targets of ProtectomiRs and confirm their association with cardioprotection in a translational pig model of acute myocardial infarction (AMI). By using a network theoretical approach, we identified 882 potential target genes of 18 previously identified protectomiRs. The Rictor gene was the most central and it was ranked first in the protectomiR-target mRNA molecular network with the highest node degree of 5. Therefore, Rictor and its targeting microRNAs were further validated in heart samples obtained from a translational pig model of AMI and cardioprotection induced by pre- or postconditioning. Three out of five Rictor-targeting pig homologue of rat ProtectomiRs showed significant upregulation in postconditioned but not in preconditioned pig hearts. Rictor was downregulated at the mRNA and protein level in ischemic postconditioning but not in ischemic preconditioning. This is the first demonstration that Rictor is the central molecular target of ProtectomiRs and that decreased Rictor expression may regulate ischemic postconditioning-, but not preconditioning-induced acute cardioprotection. We conclude that Rictor is a potential novel drug target for acute cardioprotection.


Subject(s)
MicroRNAs/metabolism , Molecular Targeted Therapy , Myocardial Infarction/metabolism , Rapamycin-Insensitive Companion of mTOR Protein/metabolism , Animals , Cardiotonic Agents , Ischemic Postconditioning , Ischemic Preconditioning, Myocardial , Rats , Swine
7.
Int J Mol Sci ; 22(19)2021 Sep 29.
Article in English | MEDLINE | ID: mdl-34638845

ABSTRACT

Helium inhalation induces cardioprotection against ischemia/reperfusion injury, the cellular mechanism of which remains not fully elucidated. Extracellular vesicles (EVs) are cell-derived, nano-sized membrane vesicles which play a role in cardioprotective mechanisms, but their function in helium conditioning (HeC) has not been studied so far. We hypothesized that HeC induces fibroblast-mediated cardioprotection via EVs. We isolated neonatal rat cardiac fibroblasts (NRCFs) and exposed them to glucose deprivation and HeC rendered by four cycles of 95% helium + 5% CO2 for 1 h, followed by 1 h under normoxic condition. After 40 h of HeC, NRCF activation was analyzed with a Western blot (WB) and migration assay. From the cell supernatant, medium extracellular vesicles (mEVs) were isolated with differential centrifugation and analyzed with WB and nanoparticle tracking analysis. The supernatant from HeC-treated NRCFs was transferred to naïve NRCFs or immortalized human umbilical vein endothelial cells (HUVEC-TERT2), and a migration and angiogenesis assay was performed. We found that HeC accelerated the migration of NRCFs and did not increase the expression of fibroblast activation markers. HeC tended to decrease mEV secretion of NRCFs, but the supernatant of HeC or the control NRCFs did not accelerate the migration of naïve NRCFs or affect the angiogenic potential of HUVEC-TERT2. In conclusion, HeC may contribute to cardioprotection by increasing fibroblast migration but not by releasing protective mEVs or soluble factors from cardiac fibroblasts.


Subject(s)
Cell Movement/drug effects , Cell-Derived Microparticles/physiology , Fibroblasts/drug effects , Helium/pharmacology , Myocardium/cytology , Animals , Animals, Newborn , Cell Line , Cell Movement/physiology , Cell-Derived Microparticles/metabolism , Cell-Derived Microparticles/ultrastructure , Cells, Cultured , Culture Media, Conditioned/pharmacology , Fibroblasts/cytology , Fibroblasts/physiology , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/physiology , Humans , Male , Microscopy, Electron, Transmission , Neovascularization, Physiologic/drug effects , Rats, Wistar
8.
Molecules ; 26(16)2021 Aug 11.
Article in English | MEDLINE | ID: mdl-34443451

ABSTRACT

Diabetes mellitus is characterized by tissue oxidative damage and impaired microcirculation, as well as worsened erythrocyte properties. Measurements of erythrocyte deformability together with determination of nitric oxide (NO) production and osmotic resistance were used for the characterization of erythrocyte functionality in lean (control) and obese Zucker diabetic fatty (ZDF) rats of two age categories. Obese ZDF rats correspond to prediabetic (younger) and diabetic (older) animals. As antioxidants were suggested to protect erythrocytes, we also investigated the potential effect of quercetin (20 mg/kg/day for 6 weeks). Erythrocyte deformability was determined by the filtration method and NO production using DAF-2DA fluorescence. For erythrocyte osmotic resistance, we used hemolytic assay. Erythrocyte deformability and NO production deteriorated during aging-both were lower in older ZDF rats than in younger ones. Three-way ANOVA indicates improved erythrocyte deformability after quercetin treatment in older obese ZDF rats only, as it was not modified or deteriorated in both (lean and obese) younger and older lean animals. NO production by erythrocytes increased post treatment in all experimental groups. Our study indicates the potential benefit of quercetin treatment on erythrocyte properties in condition of diabetes mellitus. In addition, our results suggest potential age-dependency of quercetin effects in diabetes that deserve additional research.


Subject(s)
Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/drug therapy , Erythrocytes/metabolism , Quercetin/therapeutic use , Animals , Antioxidants , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Erythrocyte Deformability/drug effects , Erythrocytes/drug effects , Lipid Metabolism/drug effects , Male , Nitric Oxide/metabolism , Osmosis , Oxidative Stress , Quercetin/pharmacology , Rats, Zucker
9.
Int J Mol Sci ; 21(5)2020 Feb 26.
Article in English | MEDLINE | ID: mdl-32111033

ABSTRACT

Quercetin (QCT) is a natural polyphenolic compound enriched in human food, mainly in vegetables, fruits and berries. QCT and its main derivatives, such as rhamnetin, rutin, hyperoside, etc., have been documented to possess many beneficial effects in the human body including their positive effects in the cardiovascular system. However, clinical implications of QCT and its derivatives are still rare. In the current paper we provide a complex picture of the most recent knowledge on the effects of QCT and its derivatives in different types of cardiac injury, mainly in ischemia-reperfusion (I/R) injury of the heart, but also in other pathologies such as anthracycline-induced cardiotoxicity or oxidative stress-induced cardiac injury, documented in in vitro and ex vivo, as well as in in vivo experimental models of cardiac injury. Moreover, we focus on cardiac effects of QCT in presence of metabolic comorbidities in addition to cardiovascular disease (CVD). Finally, we provide a short summary of clinical studies focused on cardiac effects of QCT. In general, it seems that QCT and its metabolites exert strong cardioprotective effects in a wide range of experimental models of cardiac injury, likely via their antioxidant, anti-inflammatory and molecular pathways-modulating properties; however, ageing and presence of lifestyle-related comorbidities may confound their beneficial effects in heart disease. On the other hand, due to very limited number of clinical trials focused on cardiac effects of QCT and its derivatives, clinical data are inconclusive. Thus, additional well-designed human studies including a high enough number of patients testing different concentrations of QCT are needed to reveal real therapeutic potential of QCT in CVD. Finally, several negative or controversial effects of QCT in the heart have been reported, and this should be also taken into consideration in QCT-based approaches aimed to treat CVD in humans.


Subject(s)
Cardiotonic Agents/chemistry , Cardiotonic Agents/pharmacology , Quercetin/chemistry , Quercetin/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Cardiotonic Agents/metabolism , Heart Injuries/drug therapy , Humans , Plants/chemistry , Quercetin/metabolism , Reperfusion Injury/drug therapy
10.
Int J Mol Sci ; 21(3)2020 Jan 21.
Article in English | MEDLINE | ID: mdl-31973111

ABSTRACT

The interest in non-coding RNAs, which started more than a decade ago, has still not weakened. A wealth of experimental and clinical studies has suggested the potential of non-coding RNAs, especially the short-sized microRNAs (miRs), to be used as the new generation of therapeutic targets and biomarkers of cardiovascular disease, an ever-growing public health issue in the modern world. Among the hundreds of miRs characterized so far, microRNA-1 (miR-1) and microRNA-21 (miR-21) have received some attention and have been associated with cardiac injury and cardioprotection. In this review article, we summarize the current knowledge of the function of these two miRs in the heart, their association with cardiac injury, and their potential cardioprotective roles and biomarker value. While this field has already been extensively studied, much remains to be done before research findings can be translated into clinical application for patient's benefit.


Subject(s)
Cardiotonic Agents/therapeutic use , Heart Diseases/drug therapy , MicroRNAs/therapeutic use , Animals , Antineoplastic Agents/toxicity , Arrhythmias, Cardiac/drug therapy , Biomarkers , Cardiomyopathies/drug therapy , Cardiotoxins , Cardiovascular Diseases/drug therapy , Heart , Heart Diseases/chemically induced , Heart Injuries/chemically induced , Heart Injuries/drug therapy , Humans , Radiotherapy/adverse effects
11.
Int J Mol Sci ; 21(19)2020 Oct 02.
Article in English | MEDLINE | ID: mdl-33023122

ABSTRACT

Matrix metalloproteinases (MMPs) are important in the pathogenesis of numerous diseases. The present study aimed to monitor the activation of MMP-2 and MMP-9 in spontaneously hypertensive rats (SHR) and their normotensive counterparts-Wistar-Kyoto rats (WKY). The animals were divided according to age (7, 20, and 52 weeks) and phenotype into: WKY-7, WKY-20, WKY-52, SHR-7, SHR-20 and SHR-52 groups. MMP plasma activities were determined by gelatine zymography. We monitored selected parameters of oxidative stress and antioxidant status. N-terminal pro-brain natriuretic peptide (NT-proBNP) was determined as a marker of heart function and neurohumoral activation. SHR-7 showed higher MMP-2 activity compared with WKY-7, while SHR-52 showed lower MMP-2 and MMP-9 activities compared with WKY-52. Examining age-dependent changes in MMP activities, we found a decrease in MMP-2 activity and increase in MMP-9 activity with increasing age in both phenotypes. Parameters of oxidative stress and antioxidant status as well as NT-proBNP levels were not significantly worsened due to aging in SHR. Our results suggest that hypertension is accompanied by varying MMP activation during aging. The results of our study may indicate that MMP-2 inhibition is therapeutically applicable during the development of hypertension, while in developed, stabilized and uncomplicated hypertension, systemic MMP-2 and MMP-9 inhibition may not be desirable.


Subject(s)
Aging/blood , Hypertension/blood , Matrix Metalloproteinase 2/blood , Matrix Metalloproteinase 9/blood , Age Factors , Aging/pathology , Animals , Antioxidants/metabolism , Blood Pressure , Humans , Hypertension/genetics , Hypertension/pathology , Oxidative Stress/genetics , Phenotype , Rats , Rats, Inbred SHR/blood , Rats, Inbred WKY
12.
Int J Mol Sci ; 21(21)2020 Oct 24.
Article in English | MEDLINE | ID: mdl-33114290

ABSTRACT

Iron is an essential mineral participating in different functions of the organism under physiological conditions. Numerous biological processes, such as oxygen and lipid metabolism, protein production, cellular respiration, and DNA synthesis, require the presence of iron, and mitochondria play an important role in the processes of iron metabolism. In addition to its physiological role, iron may be also involved in the adaptive processes of myocardial "conditioning". On the other hand, disorders of iron metabolism are involved in the pathological mechanisms of the most common human diseases and include a wide range of them, such as type 2 diabetes, obesity, and non-alcoholic fatty liver disease, and accelerate the development of atherosclerosis. Furthermore, iron also exerts potentially deleterious effects that may be manifested under conditions of ischemia/reperfusion (I/R) injury, myocardial infarction, heart failure, coronary artery angioplasty, or heart transplantation, due to its involvement in reactive oxygen species (ROS) production. Moreover, iron has been recently described to participate in the mechanisms of iron-dependent cell death defined as "ferroptosis". Ferroptosis is a form of regulated cell death that is distinct from apoptosis, necroptosis, and other types of cell death. Ferroptosis has been shown to be associated with I/R injury and several other cardiac diseases as a significant form of cell death in cardiomyocytes. In this review, we will discuss the role of iron in cardiovascular diseases, especially in myocardial I/R injury, and protective mechanisms stimulated by different forms of "conditioning" with a special emphasis on the novel targets for cardioprotection.


Subject(s)
Iron/metabolism , Metabolic Diseases/metabolism , Myocardial Reperfusion Injury/metabolism , Animals , Ferroptosis , Homeostasis , Humans , Metabolic Diseases/complications , Mitochondria, Heart/metabolism , Myocardial Reperfusion Injury/etiology , Reactive Oxygen Species/metabolism , Signal Transduction
13.
Int J Mol Sci ; 21(20)2020 Oct 16.
Article in English | MEDLINE | ID: mdl-33081396

ABSTRACT

Cardioprotection against ischemia/reperfusion injury is still an unmet clinical need. The transient activation of Toll-like receptors (TLRs) has been implicated in cardioprotection, which may be achieved by treatment with blood-derived extracellular vesicles (EVs). However, since the isolation of EVs from blood takes considerable effort, the aim of our study was to establish a cellular model from which cardioprotective EVs can be isolated in a well-reproducible manner. EV release was induced in HEK293 cells with calcium ionophore A23187. EVs were characterized and cytoprotection was assessed in H9c2 and AC16 cell lines. Cardioprotection afforded by EVs and its mechanism were investigated after 16 h simulated ischemia and 2 h reperfusion. The induction of HEK293 cells by calcium ionophore resulted in the release of heterogenous populations of EVs. In H9c2 and AC16 cells, stressEVs induced the downstream signaling of TLR4 and heme oxygenase 1 (HO-1) expression in H9c2 cells. StressEVs decreased necrosis due to simulated ischemia/reperfusion injury in H9c2 and AC16 cells, which was independent of TLR4 induction, but not that of HO-1. Calcium ionophore-induced EVs exert cytoprotection by inducing HO-1 in a TLR4-independent manner.


Subject(s)
Exosomes/metabolism , Heme Oxygenase-1/metabolism , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/metabolism , Animals , Calcimycin/pharmacology , Calcium Ionophores/pharmacology , Cell Death , Exosomes/drug effects , HEK293 Cells , Heme Oxygenase-1/genetics , Humans , Mice , Rats , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
14.
Molecules ; 25(15)2020 Jul 28.
Article in English | MEDLINE | ID: mdl-32731559

ABSTRACT

Search for new cardioprotective therapies is of great importance since no cardioprotective drugs are available on the market. In line with this need, several natural biomolecules have been extensively tested for their potential cardioprotective effects. Previously, we have shown that biglycan, a member of a diverse group of small leucine-rich proteoglycans, enhanced the expression of cardioprotective genes and decreased ischemia/reperfusion-induced cardiomyocyte death via a TLR-4 dependent mechanism. Therefore, in the present study we aimed to test whether decorin, a small leucine-rich proteoglycan closely related to biglycan, could exert cardiocytoprotection and to reveal possible downstream signaling pathways. Methods: Primary cardiomyocytes isolated from neonatal and adult rat hearts were treated with 0 (Vehicle), 1, 3, 10, 30 and 100 nM decorin as 20 h pretreatment and maintained throughout simulated ischemia and reperfusion (SI/R). In separate experiments, to test the mechanism of decorin-induced cardio protection, 3 nM decorin was applied in combination with inhibitors of known survival pathways, that is, the NOS inhibitor L-NAME, the PKG inhibitor KT-5823 and the TLR-4 inhibitor TAK-242, respectively. mRNA expression changes were measured after SI/R injury. Results: Cell viability of both neonatal and adult cardiomyocytes was significantly decreased due to SI/R injury. Decorin at 1, 3 and 10 nM concentrations significantly increased the survival of both neonatal and adult myocytes after SI/R. At 3nM (the most pronounced protective concentration), it had no effect on apoptotic rate of neonatal cardiac myocytes. No one of the inhibitors of survival pathways (L-NAME, KT-5823, TAK-242) influenced the cardiocytoprotective effect of decorin. MYND-type containing 19 (Zmynd19) and eukaryotic translation initiation factor 4E nuclear import factor 1 (Eif4enif1) were significantly upregulated due to the decorin treatment. In conclusion, this is the first demonstration that decorin exerts a direct cardiocytoprotective effect possibly independent of NO-cGMP-PKG and TLR-4 dependent survival signaling.


Subject(s)
Cardiotonic Agents/pharmacology , Decorin/pharmacology , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/metabolism , Signal Transduction/drug effects , Animals , Cardiotonic Agents/metabolism , Cell Survival/drug effects , Decorin/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/pathology , Rats , Rats, Wistar
15.
Molecules ; 25(1)2020 Jan 02.
Article in English | MEDLINE | ID: mdl-31906454

ABSTRACT

Background: Quercetin (QCT) was shown to exert beneficial cardiovascular effects in young healthy animals. The aim of the present study was to determine cardiovascular benefits of QCT in older, 6-month and 1-year-old Zucker diabetic fatty (ZDF) rats (model of type 2 diabetes). Methods: Lean (fa/+) and obese (fa/fa) ZDF rats of both ages were treated with QCT for 6 weeks (20 mg/kg/day). Isolated hearts were exposed to ischemia-reperfusion (I/R) injury (30 min/2 h). Endothelium-dependent vascular relaxation was measured in isolated aortas. Expression of selected proteins in heart tissue was detected by Western blotting. Results: QCT reduced systolic blood pressure in both lean and obese 6-month-old rats but had no effect in 1-year-old rats. Diabetes worsened vascular relaxation in both ages. QCT improved vascular relaxation in 6-month-old but worsened in 1-year-old obese rats and had no impact in lean controls of both ages. QCT did not exert cardioprotective effects against I/R injury and even worsened post-ischemic recovery in 1-year-old hearts. QCT up-regulated expression of eNOS in younger and PKCε expression in older rats but did not activate whole PI3K/Akt pathway. Conclusions: QCT might be beneficial for vascular function in diabetes type 2; however, increasing age and/or progression of diabetes may confound its vasculoprotective effects. QCT seems to be inefficient in preventing myocardial I/R injury in type 2 diabetes and/or higher age. Impaired activation of PI3K/Akt kinase pathway might be, at least in part, responsible for failing cardioprotection in these subjects.


Subject(s)
Blood Pressure/drug effects , Cardiovascular Diseases/drug therapy , Myocardial Ischemia/drug therapy , Myocardial Reperfusion Injury/drug therapy , Quercetin/therapeutic use , Analysis of Variance , Animals , Myocardial Ischemia/physiopathology , Myocardial Reperfusion Injury/physiopathology , Rats , Rats, Zucker , Signal Transduction/drug effects
16.
Rev Cardiovasc Med ; 20(2): 59-71, 2019 Jun 30.
Article in English | MEDLINE | ID: mdl-31344998

ABSTRACT

Remote ischemic conditioning of the heart (including pre-, per-, and post-conditioning) is a phenomenon where short episodes of non-lethal ischemia in the distant vessels within the heart or distant organs from the heart protects the myocardium against sustained ischemia/reperfusion injury. Several pathways have been proposed to be involved in the mechanisms of Remote ischemic conditioning. While triggers of Remote ischemic conditioning act in preconditioned areas, its mediators transduce protective signals via humoral or neuronal pathways to the heart. Remote ischemic conditioning is mediated via receptor and nonreceptor signaling through secondary mediators, which transfer the signal within the cardiomyocyte and activate cardioprotective pathways that lead to higher resistance of the heart to ischemia/reperfusion. Apparently, identification of endogenous signal molecules involved in the mechanisms of Remote ischemic conditioning have therapeutic implications in the management of patients suffering from myocardial ischemia through the development of diverse beneficial effects. Recently, different non-coding RNAs such as microRNAs or long non-coding RNAs have been identified as emerging factors that trigger protective mechanisms in the heart. These non-coding RNAs are transferred to the heart via extracellular vesicles that exert remote cardioprotection. This review is intended to summarize the existing knowledge about the potential role of extracellular vesicles as humoral transmitters of Remote ischemic conditioning and emphasize the involvement of non-coding RNAs in the mechanism of cardioprotection by Remote ischemic conditioning.


Subject(s)
Extracellular Vesicles/metabolism , Ischemic Postconditioning/methods , Ischemic Preconditioning/methods , Myocardial Reperfusion Injury/prevention & control , Myocardium/metabolism , RNA, Untranslated/metabolism , Animals , Extracellular Vesicles/genetics , Extracellular Vesicles/pathology , Gene Expression Regulation , Humans , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardium/pathology , RNA, Untranslated/genetics , Signal Transduction
17.
Mol Cell Biochem ; 460(1-2): 195-203, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31280435

ABSTRACT

L-Alpha-glycerylphosphorylcholine (GPC) is a widely used food supplement. GPC has been shown to exert beneficial effects in several organs; however, the cardiac effects of GPC have yet to be investigated. The aim of the present study was therefore to map out the effects of GPC on cardiac myocytes, with or without ischemia-reperfusion insult. Neonatal rat cardiac myocytes were treated with GPC at 1, 10, 80, and 100 µM concentrations for 15 min, 3 h, or 24 h, respectively. Cell viability by calcein assay and the degree of oxidative stress by DHE (superoxide level) and H2DCF (total ROS accumulation) staining were measured. In separate experiments, cardiomyocytes were pre-treated with the optimal concentration of GPC for 3 h and then cells were exposed to 4 h of simulated ischemia followed by 2 h of reperfusion (SI/R). Cell viability was measured at the end of the SI/R protocol. In normoxic conditions, the 15-min and the 3-h GPC treatment did not affect cell viability, total ROS, and superoxide levels. Under SI/R conditions, the 3-h GPC treatment protected the cardiac myocytes from SI/R-induced cell death and did not alter the level of oxidative stress. The 24-h GPC treatment in normoxic conditions resulted in significant cell death and increased oxidative stress at each concentration. Here we provide the first evidence for the cytoprotective effect of short-term GPC treatment. However, long-term administration of GPC may exert cytotoxicity in a wide concentration range in cardiac myocytes. These results may draw attention to a comprehensive cardiac safety protocol for the testing of GPC.


Subject(s)
Cytoprotection/drug effects , Glycerylphosphorylcholine/pharmacology , Myocytes, Cardiac/cytology , Animals , Animals, Newborn , Cell Death/drug effects , Cell Survival/drug effects , Glycerylphosphorylcholine/administration & dosage , Glycerylphosphorylcholine/therapeutic use , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/drug effects , Rats, Wistar
18.
Heart Fail Rev ; 23(5): 733-758, 2018 09.
Article in English | MEDLINE | ID: mdl-29862462

ABSTRACT

By virtue of their actions on NF-κB, an inflammatory nuclear transcription factor, various cytokines have been documented to play important regulatory roles in determining cardiac function under both physiological and pathophysiological conditions. Several cytokines including TNF-α, TGF-ß, and different interleukins such as IL-1 IL-4, IL-6, IL-8, and IL-18 are involved in the development of various inflammatory cardiac pathologies, namely ischemic heart disease, myocardial infarction, heart failure, and cardiomyopathies. In ischemia-related pathologies, most of the cytokines are released into the circulation and serve as biological markers of inflammation. Furthermore, there is an evidence of their direct role in the pathogenesis of ischemic injury, suggesting cytokines as potential targets for the development of some anti-ischemic therapies. On the other hand, certain cytokines such as IL-2, IL-4, IL-6, IL-8, and IL-10 are involved in the post-ischemic tissue repair and thus are considered to exert beneficial effects on cardiac function. Conflicting reports regarding the role of some cytokines in inducing cardiac dysfunction in heart failure and different types of cardiomyopathies seem to be due to differences in the nature, duration, and degree of heart disease as well as the concentrations of some cytokines in the circulation. In spite of extensive research work in this field of investigation, no satisfactory anti-cytokine therapy for improving cardiac function in any type of heart disease is available in the literature.


Subject(s)
Cytokines/metabolism , Heart Diseases/metabolism , Inflammation/metabolism , Myocardium/metabolism , Ventricular Function/physiology , Biomarkers/metabolism , Heart Diseases/physiopathology , Humans
19.
Adv Exp Med Biol ; 998: 221-235, 2017.
Article in English | MEDLINE | ID: mdl-28936743

ABSTRACT

As other stem cells, hematopoietic stem cells (HSCs) are able to produce extracellular vesicles (EVs) including exosomes and microvesicles. This chapter summarizes the knowledge about the production of EVs by the HSCs, their role in the intercellular communication, and will discuss the cargo of these EVs as well as protective effects of HSCs-derived exosomes and microvesicles in cardiovascular diseases (CVD). Available data showed that cardioprotective action of injected HSCs could not be explained by direct transdifferentiationof injected cells into the cardiomyocytes, this effect is suggested to be mediated via paracrine communication (by EVs) between donor and recipient cells. Among the cargo molecules of HSCs-derived vesicles several miRNAs, and pro-angiogenic and anti-apoptotic proteins are proposed to be the mediators of heart regeneration, mostly via neovascularization. However, the direct evidence of cardioprotective effects of HSCs-derived exosomes and microvesicles is still lacking in the literature. On the other hand, EVs produced in HSCs-derived cells, specifically dendritic cells and endothelial progenitor cells, have been shown to provide direct cardioprotective effects in CVD. Anyway, further studies are needed to be performed to assess the therapeutic potential of HSCs-derived EVs-based cardiac regenerative therapies.


Subject(s)
Cardiovascular Diseases/surgery , Exosomes/transplantation , Hematopoietic Stem Cell Transplantation , Myocardium/pathology , Myocytes, Cardiac/transplantation , Regeneration , Animals , Cardiovascular Diseases/genetics , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Exosomes/genetics , Exosomes/metabolism , Exosomes/pathology , Gene Expression Regulation , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Recovery of Function , Signal Transduction
20.
Am J Physiol Heart Circ Physiol ; 311(4): H927-H943, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27521417

ABSTRACT

Although incidence and prevalence of prediabetes are increasing, little is known about its cardiac effects. Therefore, our aim was to investigate the effect of prediabetes on cardiac function and to characterize parameters and pathways associated with deteriorated cardiac performance. Long-Evans rats were fed with either control or high-fat chow for 21 wk and treated with a single low dose (20 mg/kg) of streptozotocin at week 4 High-fat and streptozotocin treatment induced prediabetes as characterized by slightly elevated fasting blood glucose, impaired glucose and insulin tolerance, increased visceral adipose tissue and plasma leptin levels, as well as sensory neuropathy. In prediabetic animals, a mild diastolic dysfunction was observed, the number of myocardial lipid droplets increased, and left ventricular mass and wall thickness were elevated; however, no molecular sign of fibrosis or cardiac hypertrophy was shown. In prediabetes, production of reactive oxygen species was elevated in subsarcolemmal mitochondria. Expression of mitofusin-2 was increased, while the phosphorylation of phospholamban and expression of Bcl-2/adenovirus E1B 19-kDa protein-interacting protein 3 (BNIP3, a marker of mitophagy) decreased. However, expression of other markers of cardiac auto- and mitophagy, mitochondrial dynamics, inflammation, heat shock proteins, Ca2+/calmodulin-dependent protein kinase II, mammalian target of rapamycin, or apoptotic pathways were unchanged in prediabetes. This is the first comprehensive analysis of cardiac effects of prediabetes indicating that mild diastolic dysfunction and cardiac hypertrophy are multifactorial phenomena that are associated with early changes in mitophagy, cardiac lipid accumulation, and elevated oxidative stress and that prediabetes-induced oxidative stress originates from the subsarcolemmal mitochondria.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Hypertrophy, Left Ventricular/metabolism , Mitochondria, Heart/metabolism , Oxidative Stress , Prediabetic State/metabolism , Ventricular Dysfunction, Left/metabolism , Adipokines/metabolism , Adipose Tissue , Animals , Apoptosis , Autophagy , Body Composition , Calcium-Binding Proteins/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cardiomegaly/metabolism , Cardiomegaly/physiopathology , Diabetes Mellitus, Experimental/physiopathology , Diabetic Neuropathies , Diastole , Diet, High-Fat , Echocardiography , GTP Phosphohydrolases , Heat-Shock Proteins/metabolism , Hypertrophy, Left Ventricular/physiopathology , Male , Membrane Proteins/metabolism , Microscopy, Electron , Mitochondria, Heart/ultrastructure , Mitochondrial Proteins/metabolism , Mitophagy , Myocardium/metabolism , Myocardium/ultrastructure , Phosphorylation , Prediabetic State/physiopathology , Rats , Rats, Long-Evans , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Sarcolemma , TOR Serine-Threonine Kinases/metabolism , Ventricular Dysfunction, Left/physiopathology , Ventricular Pressure
SELECTION OF CITATIONS
SEARCH DETAIL