Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 525
1.
J Biochem Mol Toxicol ; 36(1): e22946, 2022 Jan.
Article En | MEDLINE | ID: mdl-34747550

Doxorubicin (DOX) is a potent antitumor agent with a broad spectrum of activity; however, irreversible cardiotoxicity resulting from DOX treatment is a major issue that limits its therapeutic use. Sirtuins (SIRTs) play an essential role in several physiological and pathological processes including oxidative stress, apoptosis, and inflammation. It has been reported that SIRT1 and SIRT3 can act as a protective molecular against DOX-induced myocardial injury through targeting numerous signaling pathways. Several natural compounds (NCs), such as resveratrol, sesamin, and berberine, with antioxidative, anti-inflammation, and antiapoptotic effects were evaluated for their potential to suppress the cardiotoxicity induced by DOX via targeting SIRT1 and SIRT3. Numerous NCs exerted their therapeutic effects on DOX-mediated cardiac damage via targeting different signaling pathways, including SIRT1/LKB1/AMPK, SIRT1/PGC-1α, SIRT1/NLRP3, and SIRT3/FoxO. SIRT3 also ameliorates cardiotoxicity by enhancing mitochondrial fusion.


Berberine/therapeutic use , Dioxoles/therapeutic use , Doxorubicin/adverse effects , Heart Diseases/enzymology , Lignans/therapeutic use , Myocardium/enzymology , Sirtuin 1/metabolism , Sirtuin 3/metabolism , Animals , Cardiotoxicity/drug therapy , Cardiotoxicity/enzymology , Doxorubicin/pharmacology , Heart Diseases/chemically induced , Heart Diseases/drug therapy , Humans
2.
Cardiovasc Res ; 118(2): 386-398, 2022 01 29.
Article En | MEDLINE | ID: mdl-33483740

Protein kinase A (PKA) is a central regulator of cardiac performance and morphology. Myocardial PKA activation is induced by a variety of hormones, neurotransmitters, and stress signals, most notably catecholamines secreted by the sympathetic nervous system. Catecholamines bind ß-adrenergic receptors to stimulate cAMP-dependent PKA activation in cardiomyocytes. Elevated PKA activity enhances Ca2+ cycling and increases cardiac muscle contractility. Dynamic control of PKA is essential for cardiac homeostasis, as dysregulation of PKA signalling is associated with a broad range of heart diseases. Specifically, abnormal PKA activation or inactivation contributes to the pathogenesis of myocardial ischaemia, hypertrophy, heart failure, as well as diabetic, takotsubo, or anthracycline cardiomyopathies. PKA may also determine sex-dependent differences in contractile function and heart disease predisposition. Here, we describe the recent advances regarding the roles of PKA in cardiac physiology and pathology, highlighting previous study limitations and future research directions. Moreover, we discuss the therapeutic strategies and molecular mechanisms associated with cardiac PKA biology. In summary, PKA could serve as a promising drug target for cardioprotection. Depending on disease types and mechanisms, therapeutic intervention may require either inhibition or activation of PKA. Therefore, specific PKA inhibitors or activators may represent valuable drug candidates for the treatment of heart diseases.


Cyclic AMP-Dependent Protein Kinases/metabolism , Heart Diseases/enzymology , Myocardial Contraction , Myocardium/enzymology , Animals , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Enzyme Activation , Heart Diseases/drug therapy , Heart Diseases/pathology , Heart Diseases/physiopathology , Humans , Molecular Targeted Therapy , Myocardium/pathology , Phosphorylation , Protein Kinase Inhibitors/therapeutic use , Signal Transduction
3.
Cardiovasc Res ; 118(2): 424-439, 2022 01 29.
Article En | MEDLINE | ID: mdl-33512477

The mechanistic target of rapamycin (mTOR) integrates several intracellular and extracellular signals involved in the regulation of anabolic and catabolic processes. mTOR assembles into two macromolecular complexes, named mTORC1 and mTORC2, which have different regulators, substrates and functions. Studies of gain- and loss-of-function animal models of mTOR signalling revealed that mTORC1/2 elicits both adaptive and maladaptive functions in the cardiovascular system. Both mTORC1 and mTORC2 are indispensable for driving cardiac development and cardiac adaption to stress, such as pressure overload. However, persistent and deregulated mTORC1 activation in the heart is detrimental during stress and contributes to the development and progression of cardiac remodelling and genetic and metabolic cardiomyopathies. In this review, we discuss the latest findings regarding the role of mTOR in the cardiovascular system, both under basal conditions and during stress, such as pressure overload, ischemia, and metabolic stress. Current data suggest that mTOR modulation may represent a potential therapeutic strategy for the treatment of cardiac diseases.


Heart Diseases/enzymology , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Myocytes, Cardiac/enzymology , TOR Serine-Threonine Kinases/metabolism , Animals , Heart Diseases/diagnosis , Heart Diseases/drug therapy , Heart Diseases/physiopathology , Humans , MTOR Inhibitors/therapeutic use , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Signal Transduction , TOR Serine-Threonine Kinases/antagonists & inhibitors , Translational Research, Biomedical
4.
Cardiovasc Toxicol ; 22(2): 152-166, 2022 02.
Article En | MEDLINE | ID: mdl-34837640

Doxorubicin (DOX) is one of the widely used anti-tumor drugs. However, DOX-induced cardiotoxicity (DIC) and hepatotoxicity (DIH) are among the side effects that limited its therapeutic efficiency and clinical applicability. This study aimed to investigate the cardioprotective and hepatoprotective potentials of curcumin (CMN)-a bioactive polyphenolic compound-in alleviating DOX-induced cardiotoxicity (DIC) and hepatotoxicity (DIH) in male rats. A single intraperitoneal (i.p.) dose of DOX (20 mg/kg) was used to induce DIC and DIH. DOX-intoxicated rats were co-treated with CMN (100 mg/kg, oral) for 10 days before and 5 days after a single dose of DOX. We studied the anti-inflammatory and anti-oxidative activities of CMN on biochemical and immunohistochemical aspects. DOX disrupted cardiac and hepatic functions and stimulated oxidative stress and inflammation in both tissues that was confirmed biochemically and immunohistochemically. DOX enhanced inflammatory interferon-gamma (IFN-γ) and upregulated immunoexpression of nuclear factor-κB (NF-κB), inducible nitric oxide synthase (iNOS), and tumor necrosis factor-alpha (TNF-α). DOX induced structural alterations in both cardiac and hepatic tissues. CMN demonstrated cardioprotective potential through reducing cardiac troponin I (cTn1) and aspartate amino transaminase (AST). In addition, CMN significantly ameliorated liver function through decreasing alanine amino transaminase (ALT) and, gamma-glutamyl transferase (GGT), total cholesterol (TC), and triglycerides (TG). CMN demonstrated anti-inflammatory potential through decreasing IFN-γ levels and immunoexpression of iNOS, NF-κB, and TNF-α. Histopathologically, CMN restored DOX-associated cardiac and liver structural alterations. CMN showed anti-oxidative and anti-inflammatory potentials in both the cardiac and hepatic tissues. In addition, cTn1, IFN-γ, and AST could be used as blood-based biomarkers.


Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Chemical and Drug Induced Liver Injury/prevention & control , Curcumin/pharmacology , Heart Diseases/prevention & control , Hepatocytes/drug effects , Myocytes, Cardiac/drug effects , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/metabolism , Oxidative Stress/drug effects , Tumor Necrosis Factor-alpha/metabolism , Animals , Cardiotoxicity , Chemical and Drug Induced Liver Injury/enzymology , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/pathology , Disease Models, Animal , Doxorubicin , Heart Diseases/chemically induced , Heart Diseases/enzymology , Heart Diseases/pathology , Hepatocytes/enzymology , Hepatocytes/pathology , Male , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Rats, Wistar , Signal Transduction
5.
Biochem Pharmacol ; 195: 114866, 2022 01.
Article En | MEDLINE | ID: mdl-34863976

Cardiovascular and heart diseases are leading causes of morbidity and mortality. Coronary artery endothelial and vascular dysfunction, inflammation, and mitochondrial dysfunction contribute to progression of heart diseases such as arrhythmias, congestive heart failure, and heart attacks. Classes of fatty acid epoxylipids and their enzymatic regulation by soluble epoxide hydrolase (sEH) have been implicated in coronary artery dysfunction, inflammation, and mitochondrial dysfunction in heart diseases. Likewise, genetic and pharmacological manipulations of epoxylipids have been demonstrated to have therapeutic benefits for heart diseases. Increasing epoxylipids reduce cardiac hypertrophy and fibrosis and improve cardiac function. Beneficial actions for epoxylipids have been demonstrated in cardiac ischemia reperfusion injury, electrical conductance abnormalities and arrhythmias, and ventricular tachycardia. This review discusses past and recent findings on the contribution of epoxylipids in heart diseases and the potential for their manipulation to treat heart attacks, arrhythmias, ventricular tachycardia, and heart failure.


Epoxide Hydrolases/metabolism , Epoxy Compounds/metabolism , Fatty Acids/metabolism , Heart Diseases/metabolism , Animals , Arrhythmias, Cardiac/drug therapy , Arrhythmias, Cardiac/enzymology , Arrhythmias, Cardiac/metabolism , Enzyme Inhibitors/therapeutic use , Epoxide Hydrolases/antagonists & inhibitors , Epoxy Compounds/chemistry , Heart Diseases/drug therapy , Heart Diseases/enzymology , Heart Failure/drug therapy , Heart Failure/enzymology , Heart Failure/metabolism , Humans , Myocardial Infarction/drug therapy , Myocardial Infarction/enzymology , Myocardial Infarction/metabolism , Solubility , Tachycardia, Ventricular/drug therapy , Tachycardia, Ventricular/enzymology , Tachycardia, Ventricular/metabolism
6.
Int J Mol Sci ; 22(21)2021 Oct 26.
Article En | MEDLINE | ID: mdl-34768967

Mitogen-activated protein kinases (MAPKs) are essential for proper cell functioning as they regulate many molecular effectors. Careful regulation of MAPKs is therefore required to avoid MAPK pathway dysfunctions and pathologies. The mammalian genome encodes about 200 phosphatases, many of which dephosphorylate the MAPKs and bring them back to an inactive state. In this review, we focus on the normal and pathological functions of dual-specificity phosphatase 9 (DUSP9)/MAP kinase phosphatases-4 (MKP-4). This cytoplasmic phosphatase, which belongs to the threonine/tyrosine dual-specific phosphatase family and was first described in 1997, is known to dephosphorylate ERK1/2, p38, JNK and ASK1, and thereby to control various MAPK pathway cascades. As a consequence, DUSP9 plays a major role in human pathologies and more specifically in cardiac dysfunction, liver metabolic syndromes, diabetes, obesity and cancer including drug response and cell stemness. Here, we recapitulate the mechanism of action of DUSP9 in the cell, its levels of regulation and its roles in the most frequent human diseases, and discuss its potential as a therapeutic target.


Dual-Specificity Phosphatases/metabolism , Mitogen-Activated Protein Kinase Phosphatases/metabolism , Animals , Dual-Specificity Phosphatases/chemistry , Dual-Specificity Phosphatases/genetics , Female , Gene Expression Regulation, Enzymologic , Heart Diseases/enzymology , Humans , MAP Kinase Signaling System , Male , Metabolic Diseases/enzymology , Metabolic Networks and Pathways , Mice , Mitogen-Activated Protein Kinase Phosphatases/chemistry , Mitogen-Activated Protein Kinase Phosphatases/genetics , Models, Molecular , Mouse Embryonic Stem Cells/enzymology , Neoplasms/enzymology , Protein Conformation , Sex Characteristics , Tissue Distribution
7.
Anesth Analg ; 133(4): 1048-1059, 2021 10 01.
Article En | MEDLINE | ID: mdl-34524989

BACKGROUND: Cardiotoxicity can be induced by the commonly used amide local anesthetic, bupivacaine. Bupivacaine can inhibit protein kinase B (AKT) phosphorylation and activated adenosine monophosphate-activated protein kinase alpha (AMPKα). It can decouple mitochondrial oxidative phosphorylation and enhance reactive oxygen species (ROS) production. Apelin enhances the phosphatidylinositol 3-kinase (PI3K)/AKT and AMPK/acetyl-CoA carboxylase (ACC) pathways, promotes the complete fatty acid oxidation in the heart, and reduces the release of ROS. In this study, we examined whether exogenous (Pyr1) apelin-13 could reverse bupivacaine-induced cardiotoxicity. METHODS: We used the bupivacaine-induced inhibition model in adult male Sprague Dawley (SD) rats (n = 48) and H9c2 cardiomyocyte cell cultures to explore the role of apelin-13 in the reversal of bupivacaine cardiotoxicity, and its possible mechanism of action. AMPKα, ACC, carnitine palmitoyl transferase (CPT), PI3K, AKT, superoxide dismutase 1 (SOD1), and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (p47-phox) were quantified. Changes in mitochondrial ultrastructure were examined, and mitochondrial DNA, cell viability, ROS release, oxygen consumption rate (OCR) were determined. RESULTS: Apelin-13 reduced bupivacaine-induced mitochondrial DNA lesions in SD rats (P < .001), while increasing the expression of AMPKα (P = .007) and PI3K (P = .002). Furthermore, apelin-13 blocked bupivacaine-induced depolarization of the mitochondrial membrane potential (P = .019) and the bupivacaine-induced increases in ROS (P = .001). Also, the AMPK pathway was activated by bupivacaine as well as apelin-13 (P = .002) in H9c2 cardiomyocytes. Additionally, the reduction in the PI3K expression by bupivacaine was mitigated by apelin-13 in H9c2 cardiomyocytes (P = .001). While the aforementioned changes induced by bupivacaine were not abated by apelin-13 after pretreatment with AMPK inhibitor compound C; the bupivacaine-induced changes were still mitigated by apelin-13, even when pretreated with PI3K inhibitor-LY294002. CONCLUSIONS: Apelin-13 treatment reduced bupivacaine-induced oxidative stress, attenuated mitochondrial morphological changes and mitochondrial DNA damage, enhanced mitochondrial energy metabolism, and ultimately reversed bupivacaine-induced cardiotoxicity. Our results suggest a role for the AMPK in apelin-13 reversal of bupivacaine-induced cardiotoxicity.


AMP-Activated Protein Kinases/metabolism , Heart Diseases/prevention & control , Intercellular Signaling Peptides and Proteins/pharmacology , Myocytes, Cardiac/drug effects , Animals , Bupivacaine , Cardiotoxicity , Cell Line , DNA Damage , Disease Models, Animal , Heart Diseases/chemically induced , Heart Diseases/enzymology , Heart Diseases/pathology , Male , Mitochondria, Heart/drug effects , Mitochondria, Heart/enzymology , Mitochondria, Heart/pathology , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Oxidative Stress , Phosphatidylinositol 3-Kinase/metabolism , Rats, Sprague-Dawley , Signal Transduction
8.
Cardiovasc Toxicol ; 21(12): 1033-1044, 2021 12.
Article En | MEDLINE | ID: mdl-34510376

Doxorubicin (DOX) is one of the most widely used chemotherapeutic drugs, but its cardiotoxicity has been shown to be a dose-restricting factor during therapy. Finding new agents for reducing these complications is still in critical need. The current study aimed to evaluate the possible cardioprotective effect of hemin (HEM) in DOX-induced cardiotoxicity and exploring the role of toll like receptor-5/nuclear factor kappa-B/tumor necrosis factor-alpha (TLR-5/NF-κB/TNF-α) and nuclear factor erythroid 2-related factor-2/hemeoxygenase-1 (Nrf-2/HO-1) signaling pathways in mediating such effect. Wistar albino rats were randomly divided into five groups. They were administered DOX by interaperitoneal (i.p.) injection (15 mg/kg) on the 5th day of the experiment with or without HEM in different doses (2.5, 5, 10 mg/kg/day) i.p. for 7 days. Results showed that the DOX group had cardiotoxicity as manifested by a significant increase in cardiac enzymes, malondialdehyde (MDA), TLR-5, NF-κB, TNF-α, and cleaved caspase-3 levels with toxic histopathological changes. Based on these findings, HEM succeeded in reducing DOX-induced cardiotoxicity in a dose-dependent effect by stimulation of Nrf-2/HO-1 and inhibition of TLR-5/NF-κB/TNF-α pathways with subsequent antioxidant, anti-inflammatory, and anti-apoptotic effects.


Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Heart Diseases/prevention & control , Heme Oxygenase (Decyclizing)/metabolism , Hemin/pharmacology , Myocytes, Cardiac/drug effects , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Toll-Like Receptor 5/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Apoptosis/drug effects , Cardiotoxicity , Disease Models, Animal , Doxorubicin , Heart Diseases/chemically induced , Heart Diseases/enzymology , Heart Diseases/pathology , Male , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Oxidative Stress/drug effects , Rats, Wistar , Signal Transduction
9.
J Cardiovasc Pharmacol ; 78(5): e690-e702, 2021 11 01.
Article En | MEDLINE | ID: mdl-34369901

ABSTRACT: This study aimed to investigate the effects of protopanaxadiol and protopanaxatriol ginsenosides on aconitine-induced cardiomyocyte injury and their regulatory mechanisms. The effects of ginsenosides on aconitine-induced cardiomyocyte damage were initially evaluated using H9c2 cells, and the molecular mechanisms were elucidated using molecular docking and western blotting. The changes in enzyme content, reactive oxygen species (ROS), calcium (Ca2+) concentration, and apoptosis were determined. Furthermore, an aconitine-induced cardiac injury rat model was established, the cardiac injury and serum physiological and biochemical indexes were measured, and the effects of ginsenoside were observed. The results showed that ginsenoside Rb1 significantly increased aconitine-induced cell viability, and its binding conformation with protein kinase B (AKT) protein was the most significant. In vitro and in vivo, Rb1 protects cardiomyocytes from aconitine-induced injury by regulating oxidative stress levels and maintaining Ca2+ concentration homeostasis. Moreover, Rb1 activated the PI3K/AKT pathway, downregulated Cleaved caspase-3 and Bax, and upregulated Bcl-2 expression. In conclusion, Rb1 protected H9c2 cells from aconitine-induced injury by maintaining Ca2+ homeostasis and activating the PI3K/AKT pathway to induce a cascade response of downstream proteins, thereby protecting cardiomyocytes from damage. These results suggested that ginsenoside Rb1 may be a potential cardiac protective drug.


Calcium/metabolism , Ginsenosides/pharmacology , Heart Diseases/prevention & control , Myocytes, Cardiac/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Sapogenins/pharmacology , Aconitine , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Cardiotoxicity , Cell Line , Disease Models, Animal , Heart Diseases/chemically induced , Heart Diseases/enzymology , Heart Diseases/pathology , Homeostasis , Male , Molecular Docking Simulation , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Oxidative Stress/drug effects , Phosphatidylinositol 3-Kinase/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Signal Transduction
10.
Int J Mol Sci ; 22(11)2021 Jun 04.
Article En | MEDLINE | ID: mdl-34199773

Despite advances in medicine, cardiac disease remains an increasing health problem associated with a high mortality rate. Maladaptive cardiac remodeling, such as cardiac hypertrophy and fibrosis, is a risk factor for heart failure; therefore, it is critical to identify new therapeutic targets. Failing heart is reported to be associated with hyper-ubiquitylation and impairment of the ubiquitin-proteasome system, indicating an importance of ubiquitylation in the development of cardiac disease. Ubiquitylation is a post-translational modification that plays a pivotal role in protein function and degradation. In 1995, homologous to E6AP C-terminus (HECT) type E3 ligases were discovered. E3 ligases are key enzymes in ubiquitylation and are classified into three families: really interesting new genes (RING), HECT, and RING-between-RINGs (RBRs). Moreover, 28 HECT-type E3 ligases have been identified in human beings. It is well conserved in evolution and is characterized by the direct attachment of ubiquitin to substrates. HECT-type E3 ligase is reported to be involved in a wide range of human diseases and health. The role of HECT-type E3 ligases in the development of cardiac diseases has been uncovered in the last decade. There are only a few review articles summarizing recent advancements regarding HECT-type E3 ligase in the field of cardiac disease. This study focused on cardiac remodeling and described the role of HECT-type E3 ligases in the development of cardiac disease. Moreover, this study revealed that the current knowledge could be exploited for the development of new clinical therapies.


Heart Diseases/enzymology , Ubiquitin-Protein Ligases/metabolism , Animals , Heart Diseases/physiopathology , Humans , Models, Cardiovascular , Stroke Volume , Ubiquitination , Vascular Remodeling
11.
Int J Mol Sci ; 22(12)2021 Jun 15.
Article En | MEDLINE | ID: mdl-34203974

In the two decades since the discovery of TNNI3K it has been implicated in multiple cardiac phenotypes and physiological processes. TNNI3K is an understudied kinase, which is mainly expressed in the heart. Human genetic variants in TNNI3K are associated with supraventricular arrhythmias, conduction disease, and cardiomyopathy. Furthermore, studies in mice implicate the gene in cardiac hypertrophy, cardiac regeneration, and recovery after ischemia/reperfusion injury. Several new papers on TNNI3K have been published since the last overview, broadening the clinical perspective of TNNI3K variants and our understanding of the underlying molecular biology. We here provide an overview of the role of TNNI3K in cardiomyopathy and arrhythmia covering both a clinical perspective and basic science advancements. In addition, we review the potential of TNNI3K as a target for clinical treatments in different cardiac diseases.


Heart Diseases/enzymology , Protein Serine-Threonine Kinases/metabolism , Animals , Disease Models, Animal , Heart Diseases/genetics , Heart Diseases/pathology , Heart Diseases/physiopathology , Humans , Molecular Targeted Therapy , Protein Serine-Threonine Kinases/genetics , Regeneration
12.
J Cardiovasc Pharmacol ; 78(1): e65-e76, 2021 07 01.
Article En | MEDLINE | ID: mdl-33929390

ABSTRACT: There is increasing evidence that angiotensin (1-7) [Ang (1-7)] is an endogenous biologically active component of the renin-angiotensin system. However, the role of the Ang (1-7)-MasR axis in postresuscitation myocardial dysfunction (PRMD) and its associated mechanism are still unclear. In this study, we investigated the effect of the Ang (1-7)-MasR axis on myocardial injury after cardiac arrest-cardiopulmonary resuscitation-restoration of spontaneous circulation. We established a model of oxygen/glucose deprivation-reperfusion in myocardial cells in vitro and a rat model of cardiac arrest-cardiopulmonary resuscitation-restoration of spontaneous circulation in vivo. The cell apoptosis rate and the expression of the superoxide anion 3-nitrotyrosine were decreased in the Ang (1-7) group in vitro and in vivo. The mean arterial pressure was decreased, whereas +LVdp/dtmax and -LVdp/dtmax were increased in rats in the Ang (1-7) group. The mRNA and protein levels of Ang II type 1 receptor, MasR, phosphoinositide 3-kinase, protein kinase B, and endothelial nitric oxide synthase were increased in the Ang (1-7) group in vivo. These results indicate that the Ang (1-7)-MasR axis can alleviate PRMD by reducing myocardial tissue damage and oxidative stress through activation of the phosphoinositide 3-kinase-protein kinase B-endothelial nitric oxide synthase signaling pathway and provide a new direction for the clinical treatment of PRMD.


Angiotensin I/pharmacology , Cardiopulmonary Resuscitation/adverse effects , Heart Arrest/therapy , Heart Diseases/prevention & control , Myocytes, Cardiac/drug effects , Nitric Oxide Synthase Type III/metabolism , Oxidative Stress/drug effects , Peptide Fragments/pharmacology , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Apoptosis/drug effects , Cells, Cultured , Disease Models, Animal , Heart Arrest/physiopathology , Heart Diseases/enzymology , Heart Diseases/etiology , Heart Diseases/physiopathology , Male , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Proto-Oncogene Mas/agonists , Proto-Oncogene Mas/genetics , Proto-Oncogene Mas/metabolism , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 2/genetics , Receptor, Angiotensin, Type 2/metabolism , Return of Spontaneous Circulation , Signal Transduction , Ventricular Function, Left/drug effects , Ventricular Pressure/drug effects
13.
J Cardiovasc Pharmacol ; 77(4): 491-500, 2021 04 01.
Article En | MEDLINE | ID: mdl-33818552

ABSTRACT: The incidence of myocardial dysfunction caused by sepsis is high, and the mortality of patients with sepsis can be significantly increased. During sepsis, oxidative stress and inflammation can lead to severe organ dysfunction. Flavone chrysin is one of the indispensable biological active ingredients for different fruits and vegetables and has antioxidant and anti-inflammatory properties. However, it is not clear whether chrysin is an effective treatment for heart dysfunction caused by sepsis. We found that it had protective effects against the harmful effects caused by LPS, manifested in improved survival, normalized cardiac function, improved partial pathological scores of myocardial tissue, and remission of apoptosis, as well as reduced oxidative stress and inflammation. Mechanism studies have found that chrysin is an important antioxidant protein, a key regulator of heme oxygenase 1 (HO-1). We found that HO-1 levels were increased after LPS intervention, and chrysin further increased HO-1 levels, along with the addition of Nrf2, a regulator of antioxidant proteins. Pretreatment with PD98059, an extracellular signal-regulated kinase-specific inhibitor, blocked chrysin-mediated phosphorylation of Nrf2 and the nuclear translocation of Nrf2. The protective effect of chrysin on sepsis-induced cardiac dysfunction was blocked by ZnPP, which is a HO-1 blocker. Chrysin increased antioxidant activity and reduced markers of oxidative stress (SOD and MDA) and inflammation (MPO and IL-1ß), all of which were blocked by ZnPP. This indicates that HO-1 is the upstream molecule regulating the protective effect of chrysin. Thus, by upregulation of HO-1, chrysin protects against LPS-induced cardiac dysfunction and inflammation by inhibiting oxidative stress.


Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Flavonoids/pharmacology , Heart Diseases/prevention & control , Heme Oxygenase-1/metabolism , Membrane Proteins/metabolism , Myocytes, Cardiac/drug effects , NF-E2-Related Factor 2/metabolism , Sepsis/drug therapy , Animals , Cell Line , Disease Models, Animal , Heart Diseases/enzymology , Heart Diseases/etiology , Heart Diseases/physiopathology , Inflammation Mediators/metabolism , Lipopolysaccharides , Male , Mice, Inbred C57BL , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Oxidative Stress/drug effects , Rats , Sepsis/chemically induced , Sepsis/enzymology , Signal Transduction , Ventricular Function, Left/drug effects
14.
Nutr Metab Cardiovasc Dis ; 31(5): 1622-1634, 2021 05 06.
Article En | MEDLINE | ID: mdl-33810953

BACKGROUND AND AIMS: It has been demonstrated that maternal low protein during development induces mitochondrial dysfunction and oxidative stress in the heart. Moderate-intensity exercise in early life, conversely, increases the overall cardiac health. Thus, we hypothesize that moderate-intensity exercise performed during young age could ameliorate the deleterious effect of maternal protein deprivation on cardiac bioenergetics. METHODS AND RESULTS: We used a rat model of maternal protein restriction during gestational and lactation period followed by an offspring treadmill moderate physical training. Pregnant rats were divided into two groups: normal nutrition receiving 17% of casein in the diet and undernutrition receiving a low-protein diet (8% casein). At 30 days of age, the male offspring were further subdivided into sedentary (NS and LS) or exercised (NT and LT) groups. Treadmill exercise was performed as follows: 4 weeks, 5 days/week, 60 min/day at 50% of maximal running capacity. Our results showed that a low-protein diet decreases oxidative metabolism and mitochondrial function associated with higher oxidative stress. In contrast, exercise rescues mitochondrial capacity and promotes a cellular resilience to oxidative stress. Up-regulation of cardiac sirtuin 1 and 3 decreased acetylation levels, redeeming from the deleterious effect of protein restriction. CONCLUSION: Our findings show that moderate daily exercise during a young age acts as a therapeutical intervention opposing the harmful effects of a maternal diet restricted in protein.


Diet, Protein-Restricted , Heart Diseases/prevention & control , Malnutrition/therapy , Mitochondria, Heart/enzymology , Oxidative Stress , Physical Conditioning, Animal , Prenatal Exposure Delayed Effects , Sirtuins/metabolism , Age Factors , Animals , Antioxidants/metabolism , Energy Metabolism , Female , Heart Diseases/enzymology , Heart Diseases/physiopathology , Male , Malnutrition/enzymology , Malnutrition/physiopathology , Maternal Nutritional Physiological Phenomena , Nutritional Status , Pregnancy , Rats, Wistar , Reactive Oxygen Species/metabolism , Running , Time Factors
15.
Cardiovasc Res ; 117(6): 1557-1566, 2021 05 25.
Article En | MEDLINE | ID: mdl-33705542

AIMS: Patients with severe respiratory syndrome caused by SARS-CoV-2 undergo cardiac complications due to hyper-inflammatory conditions. Although the presence of the virus has been detected in the myocardium of infected patients, and infection of induced pluripotent cell-derived cardiomyocytes has been demonstrated, the reported expression of Angiotensin-Converting Enzyme-2 (ACE2) in cardiac stromal cells suggests that SARS-CoV-2 may determine cardiac injury by sustaining productive infection and increasing inflammation. METHODS AND RESULTS: We analysed expression of ACE2 receptor in primary human cardiac stromal cells derived from cardiospheres, using proteomics and transcriptomics before exposing them to SARS-CoV-2 in vitro. Using conventional and high sensitivity PCR methods, we measured virus release in the cellular supernatants and monitored the intracellular viral bioprocessing. We performed high-resolution imaging to show the sites of intracellular viral production and demonstrated the presence of viral particles in the cells with electron microscopy. We finally used RT-qPCR assays to detect genes linked to innate immunity and fibrotic pathways coherently regulated in cells after exposure to the virus. CONCLUSIONS: Our findings indicate that cardiac stromal cells are susceptible to SARS-CoV-2 infection and produce variable viral yields depending on the extent of cellular ACE2 receptor expression. Interestingly, these cells also evolved towards hyper-inflammatory/pro-fibrotic phenotypes independently of ACE2 levels. Thus, SARS-CoV-2 infection of myocardial stromal cells could be involved in cardiac injury and explain the high number of complications observed in severe cases of COVID-19.


Angiotensin-Converting Enzyme 2/metabolism , COVID-19/virology , Heart Diseases/virology , Myocardium/enzymology , Receptors, Virus/metabolism , SARS-CoV-2/pathogenicity , Stromal Cells/virology , Virion/pathogenicity , Aged , Aged, 80 and over , Angiotensin-Converting Enzyme 2/genetics , Animals , COVID-19/complications , Chlorocebus aethiops , Female , Fibrosis , Heart Diseases/enzymology , Heart Diseases/pathology , Host-Pathogen Interactions , Humans , Inflammation Mediators/metabolism , Male , Middle Aged , Myocardium/ultrastructure , Phenotype , Receptors, Virus/genetics , SARS-CoV-2/ultrastructure , Spheroids, Cellular , Stromal Cells/enzymology , Stromal Cells/ultrastructure , Vero Cells , Virion/ultrastructure
16.
Cardiovasc Toxicol ; 21(4): 302-313, 2021 04.
Article En | MEDLINE | ID: mdl-33638775

Doxorubicin (DOX) is a widely used cytotoxic drug whose application is limited by its severe side effects. Little was known regarding how to offset its side effects. Therefore this study aims to explore the role of miR-200a-3p in DOX-induced cardiotoxicity and its possible mechanism. DOX-induced myocardial injury rat models were established, which were then injected with miR-200a-3p inhibitor (miR-200a-3p suppression) to observe the effects of miR-200a-3p on cell proliferation, and apoptosis. Heart function and weights of rat models were also measured. Cardiomyocytes were induced by DOX, in which PEG3 knockdown or corresponding plasmids were transfected to assess the possible effect of PEG3 on cell activity. Dual luciferase reporter assay was applied to verify the binding of PEG3 with miR-200a-3p. Elevated levels of lactate dehydrogenase (LDH), creatine kinase-MB (CK-MB) and left ventricular end-diastolic pressure (LVEDP), as well as suppressed left ventricular systolic pressure (LVSP) and ± dp/dt max were showed in myocardial injury rat models. DOX induced myocardial injury and increased miR-200a-3p expression levels. miR-200a-3p inhibitor could partially attenuate DOX-induced cardiotoxicity in rat models, while PEG3 could regulate myocardial injury in DOX-treated cell models. miR-200a-3p, by targeting PEG3 through SIRT1/NF-κB signal pathway, regulated cell proliferation, inflammation and apoptosis of myocardiocytes. The results in current study demonstrated that miR-200a-3p regulates cell proliferation and apoptosis of cardiomyocytes by targeting PEG3 through SIRT1/NF-κB signal pathway. This result may provide a potential clue for the treatment of DOX-induced cardiotoxicity.


Heart Diseases/enzymology , Inflammation Mediators/metabolism , Kruppel-Like Transcription Factors/metabolism , MicroRNAs/metabolism , Myocytes, Cardiac/enzymology , NF-kappa B/metabolism , Sirtuin 1/metabolism , Animals , Apoptosis , Cardiotoxicity , Cell Line , Cell Proliferation , Disease Models, Animal , Doxorubicin , Heart Diseases/chemically induced , Heart Diseases/genetics , Heart Diseases/pathology , Kruppel-Like Transcription Factors/genetics , Male , MicroRNAs/genetics , Myocytes, Cardiac/pathology , NF-kappa B/genetics , Rats, Wistar , Signal Transduction , Sirtuin 1/genetics , Ventricular Function, Left
17.
Inflammation ; 44(3): 1184-1193, 2021 Jun.
Article En | MEDLINE | ID: mdl-33452667

Cardiac dysfunction is a major cause leading to multiple organ failure in sepsis. Beclin-1-dependent autophagy has been evidenced to exert protective effects on hearts in sepsis. However, the mechanisms on how Beclin-1 and autophagy are regulated remains enigmatic. To explore the detailed mechanisms controlling Beclin-1-dependent autophagy in septic heart and whether melatonin could protect against sepsis via regulating cardiac autophagy, adult Sprague-Dawley (SD) rats were subjected to cecal ligation and puncture (CLP) to induce sepsis. Rats were intraperitoneally administrated with 30 mg/kg melatonin within 5-min post-CLP surgery. Our data showed that sepsis induced Becline-1 acetylation and inhibited autophagy in hearts, resulting in impaired cardiac function. However, melatonin treatment facilitated Beclin-1 deacetylation and increased autophagy in septic hearts, thus improved cardiac function. Moreover, melatonin increased the expression and activity of Sirtuin 1 (Sirt1), and inhibition of Sirt1 abolished the protective effects of melatonin on Beclin-1 deacetylation and cardiac function. In conclusion, increased Beclin-1 acetylation was involved in impaired autophagy in septic hearts, while melatonin contributed to Beclin-1 deacetylation via Sirt1, leading to improved autophagy and cardiac function in sepsis. Our study sheds light on the important role of Beclin-1 acetylation in regulating autophagy in sepsis and suggests that melatonin is a potential candidate drug for the treatment of sepsis.


Autophagy/drug effects , Beclin-1/metabolism , Heart Diseases/prevention & control , Melatonin/pharmacology , Myocytes, Cardiac/drug effects , Protein Processing, Post-Translational/drug effects , Sepsis/drug therapy , Sirtuin 1/metabolism , Ventricular Function, Left/drug effects , Acetylation , Animals , Cells, Cultured , Disease Models, Animal , Heart Diseases/enzymology , Heart Diseases/microbiology , Heart Diseases/physiopathology , Male , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Rats, Sprague-Dawley , Sepsis/enzymology , Sepsis/microbiology , Signal Transduction
18.
Cardiovasc Toxicol ; 21(3): 179-191, 2021 03.
Article En | MEDLINE | ID: mdl-33438065

Doxorubicin (DOX) is the most effective and extensively used treatment for many tumors. However, its clinical use is hampered by its cardiotoxicity. DOX-induced mitochondrial dysfunction, which causes reactive oxygen species (ROS) generation, cardiomyocyte death, bioenergetic failure, and decreased cardiac function, is a very important mechanism of cardiotoxicity. These cellular processes are all linked by mitochondrial sirtuins (SIRT3-SIRT4). Mitochondrial sirtuins preserve mitochondrial function by increasing mitochondrial metabolism, inhibiting ROS generation by activating the antioxidant enzyme manganese-dependent superoxide dismutase (MnSOD), decreasing apoptosis by activating the forkhead homeobox type O (FOXO) and P53 pathways, and increasing autophagy through AMP-activated protein kinase (AMPK)/mTOR signaling. Thus, sirtuins function at the control point of many mechanisms involved in DOX-induced cardiotoxicity. In this review, we focus on the role of mitochondrial sirtuins in mitochondrial biology and DOX-induced cardiotoxicity. A further aim is to highlight other mitochondrial processes, such as autophagy (mitophagy) and mitochondrial quality control (MQC), for which the effect of mitochondrial sirtuins on cardiotoxicity is unknown.


Antibiotics, Antineoplastic/adverse effects , Doxorubicin/adverse effects , Heart Diseases/chemically induced , Mitochondria, Heart/drug effects , Myocytes, Cardiac/drug effects , Sirtuins/metabolism , Animals , Apoptosis/drug effects , Cardiotoxicity , Energy Metabolism/drug effects , Heart Diseases/enzymology , Heart Diseases/pathology , Humans , Mitochondria, Heart/enzymology , Mitochondria, Heart/pathology , Mitophagy/drug effects , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism
19.
Clin Sci (Lond) ; 135(1): 1-17, 2021 01 15.
Article En | MEDLINE | ID: mdl-33399851

The rapid spread of the novel coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has brought into focus the key role of angiotensin-converting enzyme 2 (ACE2), which serves as a cell surface receptor required for the virus to enter cells. SARS-CoV-2 can decrease cell surface ACE2 directly by internalization of ACE2 bound to the virus and indirectly by increased ADAM17 (a disintegrin and metalloproteinase 17)-mediated shedding of ACE2. ACE2 is widely expressed in the heart, lungs, vasculature, kidney and the gastrointestinal (GI) tract, where it counteracts the deleterious effects of angiotensin II (AngII) by catalyzing the conversion of AngII into the vasodilator peptide angiotensin-(1-7) (Ang-(1-7)). The down-regulation of ACE2 by SARS-CoV-2 can be detrimental to the cardiovascular system and kidneys. Further, decreased ACE2 can cause gut dysbiosis, inflammation and potentially worsen the systemic inflammatory response and coagulopathy associated with SARS-CoV-2. This review aims to elucidate the crucial role of ACE2 both as a regulator of the renin-angiotensin system and a receptor for SARS-CoV-2 as well as the implications for Coronavirus disease 19 and its associated cardiovascular and renal complications.


Angiotensin-Converting Enzyme 2/metabolism , COVID-19/enzymology , Heart Diseases/enzymology , Kidney Diseases/enzymology , Angiotensin II/metabolism , Angiotensin-Converting Enzyme 2/genetics , Animals , COVID-19/genetics , COVID-19/metabolism , COVID-19/virology , Heart Diseases/genetics , Heart Diseases/metabolism , Heart Diseases/virology , Humans , Kidney Diseases/genetics , Kidney Diseases/metabolism , Kidney Diseases/virology , Receptors, Virus/genetics , Receptors, Virus/metabolism , Renin-Angiotensin System , SARS-CoV-2/physiology
20.
Cardiovasc Res ; 117(1): 188-200, 2021 01 01.
Article En | MEDLINE | ID: mdl-31995179

AIMS: Heart failure is a major complication in cancer treatment due to the cardiotoxic effects of anticancer drugs, especially from the anthracyclines such as doxorubicin (DXR). DXR enhances oxidative stress and stimulates matrix metalloproteinase-2 (MMP-2) in cardiomyocytes. We investigated whether MMP inhibitors protect against DXR cardiotoxicity given the role of MMP-2 in proteolyzing sarcomeric proteins in the heart and remodelling the extracellular matrix. METHODS AND RESULTS: Eight-week-old male C57BL/6J mice were treated with DXR weekly with or without MMP inhibitors doxycycline or ONO-4817 by daily oral gavage for 4 weeks. Echocardiography was used to determine cardiac function and left ventricular remodelling before and after treatment. MMP inhibitors ameliorated DXR-induced systolic and diastolic dysfunction by reducing the loss in left ventricular ejection fraction, fractional shortening, and E'/A'. MMP inhibitors attenuated adverse left ventricular remodelling, reduced cardiomyocyte dropout, and prevented myocardial fibrosis. DXR increased myocardial MMP-2 activity in part also by upregulating N-terminal truncated MMP-2. Immunogold transmission electron microscopy showed that DXR elevated MMP-2 levels within the sarcomere and mitochondria which were associated with myofilament lysis, mitochondrial degeneration, and T-tubule distention. DXR-induced myofilament lysis was associated with increased titin proteolysis in the heart which was prevented by ONO-4817. DXR also increased the level and activity of MMP-2 in human embryonic stem cell-derived cardiomyocytes, which was reduced by ONO-4817. CONCLUSIONS: MMP-2 activation is an early event in DXR cardiotoxicity and contributes to myofilament lysis by proteolyzing cardiac titin. Two orally available MMP inhibitors ameliorated DXR cardiotoxicity by attenuating intracellular and extracellular matrix remodelling, suggesting their use may be a potential prophylactic strategy to prevent heart injury during chemotherapy.


Doxycycline/pharmacology , Extracellular Matrix/drug effects , Heart Diseases/prevention & control , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase Inhibitors/pharmacology , Myocytes, Cardiac/drug effects , Phenyl Ethers/pharmacology , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects , Animals , Cardiotoxicity , Cell Line , Disease Models, Animal , Doxorubicin , Extracellular Matrix/enzymology , Extracellular Matrix/pathology , Fibrosis , Heart Diseases/chemically induced , Heart Diseases/enzymology , Heart Diseases/physiopathology , Human Embryonic Stem Cells/drug effects , Human Embryonic Stem Cells/enzymology , Humans , Male , Mice, Inbred C57BL , Mitochondria, Heart/drug effects , Mitochondria, Heart/enzymology , Mitochondria, Heart/ultrastructure , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/ultrastructure , Protein Kinases/metabolism , Proteolysis
...