Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 258
Filter
1.
Cell Biol Int ; 48(9): 1378-1391, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38922770

ABSTRACT

Oxidative stress plays a pivotal role in the development of diabetic cardiomyopathy (DCM). Previous studies have revealed that inhibition of mitochondrial fission suppressed oxidative stress and alleviated mitochondrial dysfunction and cardiac dysfunction in diabetic mice. However, no research has confirmed whether mitochondria fission accentuates hyperglycemia-induced cardiomyoblast oxidative stress through regulating fatty acid oxidation (FAO). We used H9c2 cardiomyoblasts exposed to high glucose (HG) 33 mM to simulate DCM in vitro. Excessive mitochondrial fission, poor cell viability, and lipid accumulation were observed in hyperglycemia-induced H9c2 cardiomyoblasts. Also, the cells were led to oxidative stress injury, lower adenosine triphosphate (ATP) levels, and apoptosis. Dynamin-related protein 1 (Drp1) short interfering RNA (siRNA) decreased targeted marker expression, inhibited mitochondrial fragmentation and lipid accumulation, suppressed oxidative stress, reduced cardiomyoblast apoptosis, and improved cell viability and ATP levels in HG-exposed H9c2 cardiomyoblasts, but not in carnitine palmitoyltransferase 1 (CPT1) inhibitor etomoxir treatment cells. We also found subcellular localization of CPT1 on the mitochondrial membrane, FAO, and levels of nicotinamide adenine dinucleotide phosphate (NADPH) were suppressed after exposure to HG treatment, whereas Drp1 siRNA normalized mitochondrial CPT1, FAO, and NADPH. However, the blockade of FAO with etomoxir abolished the above effects of Drp1 siRNA in hyperglycemia-induced H9c2 cardiomyoblasts. The preservation of mitochondrial function through the Drp1/CPT1/FAO pathway is the potential mechanism of inhibited mitochondria fission in attenuating oxidative stress injury of hyperglycemia-induced H9c2 cardiomyoblasts.


Subject(s)
Fatty Acids , Hyperglycemia , Mitochondrial Dynamics , Oxidation-Reduction , Oxidative Stress , Animals , Mitochondrial Dynamics/drug effects , Hyperglycemia/metabolism , Rats , Cell Line , Fatty Acids/metabolism , Apoptosis/drug effects , Cell Survival/drug effects , Dynamins/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Carnitine O-Palmitoyltransferase/metabolism , Carnitine O-Palmitoyltransferase/genetics , Diabetic Cardiomyopathies/metabolism , Diabetic Cardiomyopathies/pathology , Mitochondria/metabolism , Myoblasts, Cardiac/metabolism , Myoblasts, Cardiac/drug effects , Glucose/pharmacology , Adenosine Triphosphate/metabolism
2.
J Cardiovasc Pharmacol ; 79(1): e50-e63, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34694244

ABSTRACT

ABSTRACT: Glucagon-like peptide (GLP)-1(7-36), a major active form of GLP-1 hormone, is rapidly cleaved by dipeptidyl peptidase-4 to generate a truncated metabolite, GLP-1(9-36) which has a low affinity for GLP-1 receptor (GLP-1R). GLP-1(7-36) has been shown to have protective effects on cardiovascular system through GLP-1R-dependent pathway. Nevertheless, the cardioprotective effects of GLP-1(9-36) have not fully understood. The present study investigated the effects of GLP-1(9-36), including its underlying mechanisms against oxidative stress and apoptosis in H9c2 cells. Here, we reported that GLP-1(9-36) protects H9c2 cardiomyoblasts from hydrogen peroxide (H2O2)-induced oxidative stress by promoting the synthesis of antioxidant enzymes, glutathione peroxidase-1, catalase, and heme oxygenase-1. In addition, treatment with GLP-1(9-36) suppressed H2O2-induced apoptosis by attenuating caspase-3 activity and upregulating antiapoptotic proteins, Bcl-2 and Bcl-xL. These protective effects of GLP-1(9-36) are attenuated by blockade of PI3K-mediated Akt phosphorylation and prevention of nitric oxide synthase-induced nitric oxide production. Thus, GLP-1(9-36) represents the potential therapeutic target for prevention of oxidative stress and apoptosis in the heart via PI3K/Akt/nitric oxide synthase signaling pathway.


Subject(s)
Antioxidants , Apoptosis , Glucagon-Like Peptide 1 , Hydrogen Peroxide , Myoblasts, Cardiac , Nitric Oxide Synthase , Oxidative Stress , Phosphatidylinositol 3-Kinase , Proto-Oncogene Proteins c-akt , Animals , Rats , Antioxidants/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Cardiotoxicity , Cell Line , Glucagon-Like Peptide 1/analogs & derivatives , Glucagon-Like Peptide 1/pharmacology , Hydrogen Peroxide/toxicity , Myoblasts, Cardiac/drug effects , Myoblasts, Cardiac/enzymology , Myoblasts, Cardiac/pathology , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism , Oxidative Stress/drug effects , Phosphatidylinositol 3-Kinase/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
3.
Int J Mol Sci ; 23(4)2022 Feb 18.
Article in English | MEDLINE | ID: mdl-35216404

ABSTRACT

Tyrosine kinase inhibitors (TKIs) are associated with cardiac toxicity, which may be caused by mitochondrial toxicity. The underlying mechanisms are currently unclear and require further investigation. In the present study, we aimed to investigate in more detail the role of the enzyme complexes of the electron transfer system (ETS), mitochondrial oxidative stress, and mechanisms of cell death in cardiac toxicity associated with imatinib and sorafenib. Cardiac myoblast H9c2 cells were exposed to imatinib and sorafenib (1 to 100 µM) for 24 h. Permeabilized rat cardiac fibers were treated with both drugs for 15 min. H9c2 cells exposed to sorafenib for 24 h showed a higher membrane toxicity and ATP depletion in the presence of galactose (favoring mitochondrial metabolism) compared to glucose (favoring glycolysis) but not when exposed to imatinib. Both TKIs resulted in a higher dissipation of the mitochondrial membrane potential in galactose compared to glucose media. Imatinib inhibited Complex I (CI)- and CIII- linked respiration under both conditions. Sorafenib impaired CI-, CII-, and CIII-linked respiration in H9c2 cells cultured with glucose, whereas it inhibited all ETS complexes with galactose. In permeabilized rat cardiac myofibers, acute exposure to imatinib and sorafenib decreased CI- and CIV-linked respiration in the presence of the drugs. Electron microscopy showed enlarged mitochondria with disorganized cristae. In addition, both TKIs caused mitochondrial superoxide accumulation and decreased the cellular GSH pool. Both TKIs induced caspase 3/7 activation, suggesting apoptosis as a mechanism of cell death. Imatinib and sorafenib impaired the function of cardiac mitochondria in isolated rat cardiac fibers and in H9c2 cells at plasma concentrations reached in humans. Both imatinib and sorafenib impaired the function of enzyme complexes of the ETS, which was associated with mitochondrial ROS accumulation and cell death by apoptosis.


Subject(s)
Cardiotoxicity/etiology , Imatinib Mesylate/adverse effects , Mitochondria, Heart/drug effects , Myoblasts, Cardiac/drug effects , Myocytes, Cardiac/drug effects , Sorafenib/adverse effects , Animals , Apoptosis/drug effects , Cell Line , Electron Transport/drug effects , Glycolysis/drug effects , Membrane Potential, Mitochondrial/drug effects , Mitochondrial Membranes/drug effects , Oxidative Stress/drug effects , Rats
4.
Mol Cell Biochem ; 476(9): 3253-3260, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33886061

ABSTRACT

Pathological cardiac hypertrophy is associated with many diseases including hypertension. Recent studies have identified important roles for microRNAs (miRNAs) in many cardiac pathophysiological processes, including the regulation of cardiomyocyte hypertrophy. However, the role of miR-145-5p in the cardiac setting is still unclear. In this study, H9C2 cells were overexpressed with microRNA-145-5p, and then treated with Ang-II for 24 h, to study the effect of miR-145-5p on Ang-II-induced myocardial hypertrophy in vitro. Results showed that Ang-II treatment down-regulated miR-145-5p expression were revered after miR-145-5p overexpression. Based on results of bioinformatics algorithms, paxillin was predicted as a candidate target gene of miR-145-5p, luciferase activity assay revealed that the luciferase activity of cells was substantial downregulated the following co-transfection with wild paxillin 3'UTR and miR-145-5p compared to that in scramble control, while the inhibitory effect of miR-145-5p was abolished after transfection of mutant paxillin 3'UTR. Additionally, overexpression of miR-145-5p markedly inhibited activation of Rac-1/ JNK /c-jun/ NFATc3 and ANP expression and induced SIRT1 expression in Ang-II treated H9c2 cells. Jointly, our study suggested that miR-145-5p inhibited cardiac hypertrophy by targeting paxillin and through modulating Rac-1/ JNK /c-jun/ NFATc3/ ANP / Sirt1 signaling, therefore proving novel downstream molecular pathway of miR-145-5p in cardiac hypertrophy.


Subject(s)
Angiotensin II/toxicity , Cardiomegaly/drug therapy , Gene Expression Regulation/drug effects , MicroRNAs/genetics , Myoblasts, Cardiac/drug effects , Paxillin/antagonists & inhibitors , Animals , Atrial Natriuretic Factor/genetics , Atrial Natriuretic Factor/metabolism , Cardiomegaly/chemically induced , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cells, Cultured , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , Myoblasts, Cardiac/metabolism , Myoblasts, Cardiac/pathology , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , Rats , Sirtuin 1/genetics , Sirtuin 1/metabolism , Vasoconstrictor Agents/toxicity , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism
5.
Nature ; 525(7570): 479-85, 2015 Sep 24.
Article in English | MEDLINE | ID: mdl-26375005

ABSTRACT

The elucidation of factors that activate the regeneration of the adult mammalian heart is of major scientific and therapeutic importance. Here we found that epicardial cells contain a potent cardiogenic activity identified as follistatin-like 1 (Fstl1). Epicardial Fstl1 declines following myocardial infarction and is replaced by myocardial expression. Myocardial Fstl1 does not promote regeneration, either basally or upon transgenic overexpression. Application of the human Fstl1 protein (FSTL1) via an epicardial patch stimulates cell cycle entry and division of pre-existing cardiomyocytes, improving cardiac function and survival in mouse and swine models of myocardial infarction. The data suggest that the loss of epicardial FSTL1 is a maladaptive response to injury, and that its restoration would be an effective way to reverse myocardial death and remodelling following myocardial infarction in humans.


Subject(s)
Follistatin-Related Proteins/metabolism , Myocardium/metabolism , Pericardium/growth & development , Pericardium/metabolism , Regeneration , Animals , Cell Cycle/drug effects , Cell Proliferation/drug effects , Culture Media, Conditioned/pharmacology , Female , Follistatin-Related Proteins/genetics , Humans , Male , Mice , Myoblasts, Cardiac/cytology , Myoblasts, Cardiac/drug effects , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Pericardium/cytology , Pericardium/drug effects , Rats , Regeneration/drug effects , Signal Transduction , Swine , Transgenes/genetics
6.
Lipids Health Dis ; 20(1): 151, 2021 Nov 02.
Article in English | MEDLINE | ID: mdl-34727932

ABSTRACT

BACKGROUND: Acylcarnitine is an intermediate product of fatty acid oxidation. It is reported to be closely associated with the occurrence of diabetic cardiomyopathy (DCM). However, the mechanism of acylcarnitine affecting myocardial disorders is yet to be explored. This current research explores the different chain lengths of acylcarnitines as biomarkers for the early diagnosis of DCM and the mechanism of acylcarnitines for the development of DCM in-vitro. METHODS: In a retrospective non-interventional study, 50 simple type 2 diabetes mellitus patients and 50 DCM patients were recruited. Plasma samples from both groups were analyzed by high throughput metabolomics and cluster heat map using mass spectrometry. Principal component analysis was used to compare the changes occurring in the studied 25 acylcarnitines. Multivariable binary logistic regression was used to analyze the odds ratio of each group for factors and the 95% confidence interval in DCM. Myristoylcarnitine (C14) exogenous intervention was given to H9c2 cells to verify the expression of lipid metabolism-related protein, inflammation-related protein expression, apoptosis-related protein expression, and cardiomyocyte hypertrophy and fibrosis-related protein expression. RESULTS: Factor 1 (C14, lauroylcarnitine, tetradecanoyldiacylcarnitine, 3-hydroxyl-tetradecanoylcarnitine, arachidic carnitine, octadecanoylcarnitine, 3-hydroxypalmitoleylcarnitine) and factor 4 (octanoylcarnitine, hexanoylcarnitine, decanoylcarnitine) were positively correlated with the risk of DCM. Exogenous C14 supplementation to cardiomyocytes led to increased lipid deposition in cardiomyocytes along with the obstacles in adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) signaling pathways and affecting fatty acid oxidation. This further caused myocardial lipotoxicity, ultimately leading to cardiomyocyte hypertrophy, fibrotic remodeling, and increased apoptosis. However, this effect was mitigated by the AMPK agonist acadesine. CONCLUSIONS: The increased plasma levels in medium and long-chain acylcarnitine extracted from factors 1 and 4 are closely related to the risk of DCM, indicating that these factors can be an important tool for DCM risk assessment. C14 supplementation associated lipid accumulation by inhibiting the AMPK/ACC/CPT1 signaling pathway, aggravated myocardial lipotoxicity, increased apoptosis apart from cardiomyocyte hypertrophy and fibrosis were alleviated by the acadesine.


Subject(s)
Carnitine/analogs & derivatives , Diabetes Mellitus, Type 2/complications , Diabetic Cardiomyopathies/metabolism , Lipid Metabolism , Adult , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Biomarkers/blood , Carnitine/blood , Carnitine/chemistry , Carnitine/pharmacology , Cell Line , Diabetes Mellitus, Type 2/metabolism , Female , Humans , Lipid Metabolism/drug effects , Male , Mass Spectrometry , Middle Aged , Myoblasts, Cardiac/drug effects , Myoblasts, Cardiac/metabolism , Myristic Acids/pharmacology , Rats , Retrospective Studies , Ribonucleosides/pharmacology , Risk Factors
7.
Int J Mol Sci ; 22(10)2021 May 11.
Article in English | MEDLINE | ID: mdl-34064664

ABSTRACT

Rutin is a flavonoid with antioxidant property. It has been shown to exert cardioprotection against cardiomyocyte hypertrophy. However, studies regarding its antihypertrophic property are still lacking, whether it demonstrates similar antihypertrophic effect to its metabolite, quercetin. Hence, this study aimed to investigate the effects of both flavonoids on oxidative stress and mitogen-activated protein kinase (MAPK) pathway in H9c2 cardiomyocytes that were exposed to angiotensin II (Ang II) to induce hypertrophy. Cardiomyocytes were exposed to Ang II (600 nM) with or without quercetin (331 µM) or rutin (50 µM) for 24 h. A group given vehicle served as the control. The concentration of the flavonoids was chosen based on the reported effective concentration to reduce cell hypertrophy or cardiac injury in H9c2 cells. Exposure to Ang II increased cell surface area, intracellular superoxide anion level, NADPH oxidase and inducible nitric oxide synthase activities, and reduced cellular superoxide dismutase activity and nitrite level, which were similarly reversed by both rutin and quercetin. Rutin had no significant effects on phosphorylated proteins of extracellular signal-related kinases (ERK1/2) and p38 but downregulated phosphorylated c-Jun N-terminal kinases (JNK1/2), which were induced by Ang II. Quercetin, on the other hand, had significantly downregulated the phosphorylated proteins of ERK1/2, p38, and JNK1/2. The quercetin inhibitory effect on JNK1/2 was stronger than the rutin. In conclusion, both flavonoids afford similar protective effects against Ang II-induced cardiomyocyte hypertrophy, but they differently modulate MAPK pathway.


Subject(s)
Angiotensin II/toxicity , Gene Expression Regulation, Enzymologic/drug effects , Hypertrophy/metabolism , Mitogen-Activated Protein Kinases/metabolism , Myoblasts, Cardiac/metabolism , Quercetin/pharmacology , Rutin/pharmacology , Animals , Antioxidants/pharmacology , Cells, Cultured , Hypertrophy/chemically induced , Hypertrophy/drug therapy , Hypertrophy/pathology , Mitogen-Activated Protein Kinases/genetics , Myoblasts, Cardiac/cytology , Myoblasts, Cardiac/drug effects , NADPH Oxidases/metabolism , Nitric Oxide/metabolism , Phosphorylation , Rats , Reactive Oxygen Species/metabolism , Vasoconstrictor Agents/toxicity
8.
Int J Mol Sci ; 22(6)2021 Mar 23.
Article in English | MEDLINE | ID: mdl-33806909

ABSTRACT

Kirenol (KRL) is a biologically active substance extracted from Herba Siegesbeckiae. This natural type of diterpenoid has been widely adopted for its important anti-inflammatory and anti-rheumatic properties. Despite several studies claiming the benefits of KRL, its cardiac effects have not yet been clarified. Cardiotoxicity remains a key concern associated with the long-term administration of doxorubicin (DOX). The generation of reactive oxygen species (ROS) causes oxidative stress, significantly contributing to DOX-induced cardiac damage. The purpose of the current study is to investigate the cardio-protective effects of KRL against apoptosis in H9c2 cells induced by DOX. The analysis of cellular apoptosis was performed using the terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) staining assay and measuring the modulation in the expression levels of proteins involved in apoptosis and Nrf2 signaling, the oxidative stress markers. Furthermore, Western blotting was used to determine cell survival. KRL treatment, with Nrf2 upregulation and activation, accompanied by activation of PI3K/AKT, could prevent the administration of DOX to induce cardiac oxidative stress, remodeling, and other effects. Additionally, the diterpenoid enhanced the activation of Bcl2 and Bcl-xL, while suppressing apoptosis marker proteins. As a result, KRL is considered a potential agent against hypertrophy resulting from cardiac deterioration. The study results show that KRL not only activates the IGF-IR-dependent p-PI3K/p-AKT and Nrf2 signaling pathway, but also suppresses caspase-dependent apoptosis.


Subject(s)
Cardiotonic Agents/pharmacology , Diterpenes/pharmacology , NF-E2-Related Factor 2/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Cell Death/drug effects , Cell Line , Cell Survival/drug effects , Cytoskeleton/metabolism , Diterpenes/chemistry , Doxorubicin/adverse effects , Humans , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Myoblasts, Cardiac/drug effects , Myoblasts, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Natriuretic Peptides/metabolism , Phosphorylation , Protein Transport
9.
Molecules ; 26(23)2021 Dec 01.
Article in English | MEDLINE | ID: mdl-34885888

ABSTRACT

Although numerous studies have demonstrated the biological and multifaceted nature of dimethyl sulfoxide (DMSO) across different in vitro models, the direct effect of "non-toxic" low DMSO doses on cardiac and cancer cells has not been clearly explored. In the present study, H9c2 cardiomyoblasts and MCF-7 breast cancer cells were treated with varying concentrations of DMSO (0.001-3.7%) for 6 days. Here, DMSO doses < 0.5% enhanced the cardiomyoblasts respiratory control ratio and cellular viability relative to the control cells. However, 3.7% DMSO exposure enhanced the rate of apoptosis, which was driven by mitochondrial dysfunction and oxidative stress in the cardiomyoblasts. Additionally, in the cancer cells, DMSO (≥0.009) led to a reduction in the cell's maximal respiratory capacity and ATP-linked respiration and turnover. As a result, the reduced bioenergetics accelerated ROS production whilst increasing early and late apoptosis in these cells. Surprisingly, 0.001% DMSO exposure led to a significant increase in the cancer cells proliferative activity. The latter, therefore, suggests that the use of DMSO, as a solvent or therapeutic compound, should be applied with caution in the cancer cells. Paradoxically, in the cardiomyoblasts, the application of DMSO (≤0.5%) demonstrated no cytotoxic or overt therapeutic benefits.


Subject(s)
Apoptosis/drug effects , Dimethyl Sulfoxide/pharmacology , Mitochondria/drug effects , Oxidative Stress/drug effects , Cell Line , Cell Survival/drug effects , Energy Metabolism/drug effects , Humans , MCF-7 Cells , Mitochondria/metabolism , Myoblasts, Cardiac/cytology , Myoblasts, Cardiac/drug effects , Reactive Oxygen Species/metabolism
10.
Proc Natl Acad Sci U S A ; 114(23): E4582-E4591, 2017 06 06.
Article in English | MEDLINE | ID: mdl-28526717

ABSTRACT

The injury phase after myocardial infarcts occurs during reperfusion and is a consequence of calcium release from internal stores combined with calcium entry, leading to cell death by apoptopic and necrotic processes. The mechanism(s) by which calcium enters cells has(ve) not been identified. Here, we identify canonical transient receptor potential channels (TRPC) 3 and 6 as the cation channels through which most of the damaging calcium enters cells to trigger their death, and we describe mechanisms activated during the injury phase. Working in vitro with H9c2 cardiomyoblasts subjected to 9-h hypoxia followed by 6-h reoxygenation (H/R), and analyzing changes occurring in areas-at-risk (AARs) of murine hearts subjected to a 30-min ischemia followed by 24-h reperfusion (I/R) protocol, we found: (i) that blocking TRPC with SKF96365 significantly ameliorated damage induced by H/R, including development of the mitochondrial permeability transition and proapoptotic changes in Bcl2/BAX ratios; and (ii) that AAR tissues had increased TUNEL+ cells, augmented Bcl2/BAX ratios, and increased p(S240)NFATc3, p(S473)AKT, p(S9)GSK3ß, and TRPC3 and -6 proteins, consistent with activation of a positive-feedback loop in which calcium entering through TRPCs activates calcineurin-mediated NFATc3-directed transcription of TRPC genes, leading to more Ca2+ entry. All these changes were markedly reduced in mice lacking TRPC3, -6, and -7. The changes caused by I/R in AAR tissues were matched by those seen after H/R in cardiomyoblasts in all aspects except for p-AKT and p-GSK3ß, which were decreased after H/R in cardiomyoblasts instead of increased. TRPC should be promising targets for pharmacologic intervention after cardiac infarcts.


Subject(s)
Cell Hypoxia/physiology , Myocardial Reperfusion Injury/etiology , TRPC Cation Channels/metabolism , Animals , Apoptosis , Calcium Channel Blockers/pharmacology , Calcium Signaling , Cell Hypoxia/drug effects , Cell Line , Disease Models, Animal , Imidazoles/pharmacology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Models, Cardiovascular , Myoblasts, Cardiac/drug effects , Myoblasts, Cardiac/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Signal Transduction , TRPC Cation Channels/deficiency , TRPC Cation Channels/genetics , TRPC6 Cation Channel
11.
Environ Toxicol ; 35(10): 1043-1049, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32415908

ABSTRACT

In this study, we used ICI 182 780 (ICI), an estrogen receptor (ER) antagonist, to investigate the estrogenic activity of Danshen, and to further explored whether Danshen extract can block Leu27IGF-II-induced hypertrophy in H9c2 cardiomyoblast cells. We first used an IGF-II analog Leu27IGF-II, which specifically activates IGF2R signaling cascades and induces H9c2 cardiomyoblast cell hypertrophy. However, Danshen extract completely inhibited Leu27IGF-II-induced cell size increase, ANP and BNP hypertrophic marker expression, and IGF2R induction. We also observed that Danshen extract inhibited calcineurin protein expression and NFAT3 nuclear translocation, leading to suppression of Leu27IGF-II-induced cardiac hypertrophy. Moreover, the anti-Leu27IGF-II-IGF2R signaling effect of Danshen was totally reversed by ICI, which suggest the cardio protective effect of Danshen is mediated through estrogen receptors. Our study suggests that, Danshen exerts estrogenic activity, and thus, it could be used as a selective ER modulator in IGFIIR induced hypertrophy model.


Subject(s)
Cell Enlargement/drug effects , Drugs, Chinese Herbal/pharmacology , Insulin-Like Growth Factor II/analogs & derivatives , Myoblasts, Cardiac/drug effects , Receptor, IGF Type 2/metabolism , Salvia miltiorrhiza/chemistry , Animals , Calcineurin/metabolism , Cardiomegaly/prevention & control , Cell Line , Cell Survival/drug effects , Drugs, Chinese Herbal/isolation & purification , Estrogen Receptor Antagonists/pharmacology , Fulvestrant/pharmacology , Insulin-Like Growth Factor II/pharmacology , Myoblasts, Cardiac/metabolism , Myoblasts, Cardiac/pathology , Protein Transport , Rats , Receptors, Estrogen/metabolism , Signal Transduction
12.
Molecules ; 25(22)2020 Nov 11.
Article in English | MEDLINE | ID: mdl-33187371

ABSTRACT

Natural products black cumin-Nigella sativa (N. sativa) and wild garlic-Allium ursinum (AU) are known for their potential role in reducing cardiovascular risk factors, including antracycline chemotherapy. Therefore, this study investigates the effect of N. sativa and AU water and methanolic extracts in a cellular model of doxorubicin (doxo)-induced cardiotoxicity. The extracts were characterized using Ultraviolet-visible (UV-VIS) spectroscopy, Fourier-transform infrared (FT-IR) spectroscopy, Liquid Chromatography coupled with Mass Spectrometry (LC-MS) and Gas Chromatography coupled with Mass Spectrometry (GC-MS) techniques. Antioxidant activity was evaluated on H9c2 cells. Cytosolic and mitochondrial reactive oxygen species (ROS) release was evaluated using 2',7'-dichlorofluorescin-diacetate (DHCF-DA) and mitochondria-targeted superoxide indicator (MitoSOX red), respectively. Mitochondrial membrane depolarization was evaluated by flow cytometry. LC-MS analysis identified 12 and 10 phenolic compounds in NSS and AU extracts, respectively, with flavonols as predominant compounds. FT-IR analysis identified the presence of carbohydrates, amino acids and lipids in both plants. GC-MS identified the sulfur compounds in the AU water extract. N. sativa seeds (NSS) methanolic extract had the highest antioxidant activity reducing both intracellular and mitochondrial ROS release. All extracts (excepting AU methanolic extract) preserved H9c2 cells viability. None of the investigated plants affected the mitochondrial membrane depolarization. N. sativa and AU are important sources of bioactive compounds with increased antioxidant activities, requiring different extraction solvents to obtain the pharmacological effects.


Subject(s)
Allium/chemistry , Antioxidants/chemistry , Doxorubicin/chemistry , Myoblasts, Cardiac/drug effects , Nigella sativa/chemistry , Plant Extracts/pharmacology , Animals , Cardiotoxicity , Cell Line , Cell Survival , Flavonols/analysis , Gas Chromatography-Mass Spectrometry , Membrane Potential, Mitochondrial , Phenols/pharmacology , Polyphenols/chemistry , Rats , Reactive Oxygen Species/metabolism , Risk Factors , Seeds/chemistry , Spectrophotometry, Ultraviolet , Spectroscopy, Fourier Transform Infrared
13.
Cell Biol Toxicol ; 35(2): 111-127, 2019 04.
Article in English | MEDLINE | ID: mdl-30006751

ABSTRACT

Cigarette smoking causes a vast array of diseases including cardiovascular diseases. Our laboratory focuses on investigating cigarette smoke (CS)-induced cardiovascular malfunction and the responsible mechanisms utilizing the model, c-kit-positive cardiac stem cells (CSCs). The main objective of our study is to investigate whether CS extracts (CSEs) cause impairment of CSC functions via oxidative damage. We hypothesized that CSE, via oxidative modifications of CSC proteins and antioxidant enzymes, can modulate CSC functions and these modifications can be attenuated by ascorbate treatment. Our specific aims are (1) to investigate CSE-induced oxidative modification of CSC proteins via carbonylation, and prevention by ascorbic acid; (2) to investigate CSE-induced oxidative modification of antioxidant enzymes and ascorbic acid-mediated modulations; and (3) to investigate CSE-induced changes in CSC functions and protection by ascorbic acid. CSCs were cultured, and the aqueous extracts of CSE were prepared. CSE-induced modulations of CSC viability, oxidative modification of proteins, and antioxidant enzyme activities were detected using standard assays including Apostain, bromodeoxyuridine, and Oxiblot. CSE caused oxidative modification of CSC proteins, changed antioxidant enzyme levels, attenuated CSC proliferation, and accelerated CSC apoptosis. Ascorbic acid prevented CSE-induced CSC malfunctions, and ascorbic acid therapy might be useful in smoker CSC recipients and to condition CSCs prior to the transplant in the future. Cardiac stem cell therapy is currently undergoing in clinical trials.


Subject(s)
Ascorbic Acid/pharmacology , Myoblasts, Cardiac/drug effects , Oxidative Stress/drug effects , Smoke/adverse effects , Vitamins/pharmacology , Animals , Cardiomyopathies/etiology , Cardiomyopathies/prevention & control , Cells, Cultured , Myoblasts, Cardiac/cytology , Rats , Rats, Inbred F344 , Tobacco Smoke Pollution/adverse effects
14.
Environ Toxicol ; 34(8): 921-927, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31066208

ABSTRACT

Hyperkalemia is often associated with cardiac dysfunction. In this study an earthworm extract (dilong) was prepared from dried Pheretima aspergillum powder and its effect against high-KCl challenge was determined in H9c2 cardiomyoblast cells. H9c2 cells pre-treated with dilong (31.25, 62.5, 125, and 250 mg/mL) for 24 hours, where challenged with different doses of KCl treatment for 3 hours to determine the protective mechanisms of dilong against cardiac fibrosis. High-KCl administration induced mitochondrial injury and elevated the levels of pro-apoptotic proteins. The mediators of fibrosis such as ERK, uPA, SP1, and CTGF were also found to be upregulated in high-KCl condition. However, dilong treatment enhanced IGF1R/PI3k/Akt activation which is associated with cell survival. In addition, dilong also reversed high-KCl induced cardiac fibrosis related events in H9c2 cells and displayed a strong cardio-protective effect. Therefore, dilong is a potential agent to overcome cardiac events associated with high-KCl toxicity.


Subject(s)
Mitochondria, Heart/drug effects , Myoblasts, Cardiac/drug effects , Oligochaeta , Potassium Chloride/toxicity , Animals , Apoptosis Regulatory Proteins/metabolism , Cell Line , Cell Survival , Fibrosis , Myoblasts, Cardiac/pathology , Protective Agents/pharmacology
15.
J Asian Nat Prod Res ; 21(5): 476-493, 2019 May.
Article in English | MEDLINE | ID: mdl-29322879

ABSTRACT

Diabetic cardiomyopathy (DCM) is one of the chief diabetes mellitus complications. Inflammation factors may be one reason for the damage from DM. The purpose of this research is to study the potential protective effects of puerarin on DM and the possible mechanisms of action related to NF-κB signal pathway. Following administration of puerarin to the disease model rat, several changes were observed including the changes of serum biochemical index, improved diastolic dysfunction, and enhanced endogenous antioxidant enzymes activities, further NF-κB signaling activation. Puerarin showed cardio-protective effects on DCM by inhibiting inflammation, and it might be a potential candidate for the treatment of DCM.


Subject(s)
Diabetes Mellitus, Experimental/complications , Diabetic Cardiomyopathies/prevention & control , Inflammation/drug therapy , Isoflavones/pharmacology , Animals , Cell Line , Cells, Cultured , Glucose/toxicity , Isoflavones/chemistry , Molecular Structure , Myoblasts, Cardiac/drug effects , Myocardium/cytology , NF-kappa B , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
16.
J Cell Biochem ; 119(9): 7855-7864, 2018 09.
Article in English | MEDLINE | ID: mdl-29932238

ABSTRACT

Aberrant expression of leucine zipper- and sterile ɑ motif-containing kinase (ZAK) observed in pathological human myocardial tissue is associated with the progression and elevation of hypertrophy. Our previous reports have correlated high levels of estrogen (E2) and abundant estrogen receptor (ER) α with a low incidence of pathological cardiac-hypertrophy and heart failure in the premenopause female population. However, the effect of elevated ERß expression is not well known yet. Therefore, in this study, we have analyzed the cardioprotective effects and mechanisms of E2 and/or ERß against ZAK overexpression-induced cellular hypertrophy. We have used transient transfection to overexpress ERß into the ZAK tet-on H9c2 cells that harbor the doxycycline-inducible ZAK plasmid. The results show that ZAK overexpression in H9c2 cells resulted in hypertrophic effects, which was correlated with the upregulation of p-JNK and p-p38 MAPKs and their downstream transcription factors c-Jun and GATA-4. However, ERß and E2 with ERß overexpressions totally suppressed the effects of ZAK overexpression and inhibited the levels of p-JNK, p-p38, c-Jun, and GATA-4 effectively. Our results further reveal that ERß directly binds with ZAK under normal conditions; however, ZAK overexpression reduced the association of ZAK-ERß. Interestingly, increase in ERß and E2 along with ERß overexpression both enhanced the binding strengths of ERß and ZAK and reduced the ZAK protein level. ERß overexpression also suppressed the E3 ligase-casitas B-lineage lymphoma (CBL) and attenuated CBL-phosphoinositide 3-kinase (PI3K) protein association to prevent PI3K protein degradation. Moreover, ERß and/or E2 blocked ZAK nuclear translocation via the inhibition of small ubiquitin-like modifier (SUMO)-1 modification. Taken together, our results further suggest that ERß overexpression strongly suppresses ZAK-induced cellular hypertrophy and myocardial damage.


Subject(s)
Estrogen Receptor beta/genetics , Myoblasts, Cardiac/cytology , Protein Kinases/metabolism , SUMO-1 Protein/metabolism , Animals , Cell Enlargement , Cell Line , Estrogen Receptor beta/metabolism , Estrogens/pharmacology , Gene Expression Regulation , Myoblasts, Cardiac/drug effects , Myoblasts, Cardiac/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinases/genetics , Proto-Oncogene Proteins c-cbl , Rats
17.
J Cell Biochem ; 119(9): 7678-7686, 2018 09.
Article in English | MEDLINE | ID: mdl-29923625

ABSTRACT

Patients affected by long-chain 3-hydroxyacyl-CoA dehydrogenase (LCHAD) deficiency predominantly present severe liver and cardiac dysfunction, as well as neurological symptoms during metabolic crises, whose pathogenesis is still poorly known. In this study, we demonstrate for the first time that pathological concentrations of 3-hydroxypalmitic acid (3HPA), the long-chain hydroxyl fatty acid (LCHFA) that most accumulates in LCHAD deficiency, significantly decreased adenosine triphosphate-linked and uncoupled mitochondrial respiration in intact cell systems consisting of heart fibers, cardiomyocytes, and hepatocytes, but less intense in diced forebrain. 3HPA also significantly reduced mitochondrial Ca2+ retention capacity and membrane potential in Ca2+ -loaded mitochondria more markedly in the heart and the liver, with mild or no effects in the brain, supporting a higher susceptibility of the heart and the liver to the toxic effects of this fatty acid. It is postulated that disruption of mitochondrial energy and Ca2+ homeostasis caused by the accumulation of LCHFA may contribute toward the severe cardiac and hepatic clinical manifestations observed in the affected patients.


Subject(s)
Hepatocytes/metabolism , Mitochondria/drug effects , Myoblasts, Cardiac/metabolism , Palmitic Acids/adverse effects , Adenosine Triphosphate/metabolism , Animals , Brain/cytology , Brain/drug effects , Brain/metabolism , Calcium/metabolism , Cell Line , Hep G2 Cells , Hepatocytes/cytology , Hepatocytes/drug effects , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Myoblasts, Cardiac/cytology , Myoblasts, Cardiac/drug effects , Rats , Rats, Wistar
18.
Cell Physiol Biochem ; 46(5): 2031-2040, 2018.
Article in English | MEDLINE | ID: mdl-29723857

ABSTRACT

BACKGROUND/AIMS: Diabetic cardiomyopathy is associated with increased apoptosis and suppressed autophagy in cardiac cells. The polyphenol resveratrol has shown beneficial effects in various cardiovascular diseases. This study investigated if resveratrol protected cardiac cells by modulating apoptosis and autophagy in the context of diabetes. METHODS: H9c2 cardiac myoblast cells were exposed to high glucose combined with palmitate. Autophagy was evaluated by estimating LC3-II/I ratio, P62 protein levels, and LC3 fluorescent puncta. Apoptosis was assessed by using terminal deoxynucleotidyl transferase dUTP nick end labelling (TUNEL), flow cytometry, and analysis of the protein expression of apoptotic markers (cleavage of caspase-3 and PARP). RESULTS: High glucose and palmitate suppressed autophagic activity and exacerbated apoptotic cell death in cardiac myoblast cells. Resveratrol restored autophagy and attenuated apoptosis in cells upon diabetic stimuli. Moreover, resveratrol activated AMPK and JNK1, thereby suppressing mTOR and its downstream effectors p70S6K1 and 4EBP1, as well as disrupting the Beclin1-Bcl-2 complex. CONCLUSION: Resveratrol protects cardiac cells by regulating the switch between autophagy and apoptotic machinery under diabetic conditions, which is attributed by AMPK-mediated phosphorylation of mTORC1/p70S6K1/4EBP1 and JNK-mediated dissociation of Beclin1-Bcl-2. Our study suggests that autophagy may be an important target for resveratrol in the treatment of diabetic cardiomyopathy.


Subject(s)
Antioxidants/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Glucose/metabolism , Myoblasts, Cardiac/drug effects , Palmitates/metabolism , Stilbenes/pharmacology , Animals , Cell Line , Diabetic Cardiomyopathies/drug therapy , Diabetic Cardiomyopathies/metabolism , Hyperglycemia/drug therapy , Hyperglycemia/metabolism , Myoblasts, Cardiac/cytology , Myoblasts, Cardiac/metabolism , Rats , Resveratrol
19.
Biomacromolecules ; 19(5): 1646-1662, 2018 05 14.
Article in English | MEDLINE | ID: mdl-29596739

ABSTRACT

A dual cross-linking strategy was developed to answer the urgent need for fatigue-resistant, cytocompatible, and in situ forming tough hydrogels. Clickable, yet calcium-binding derivatives of alginate were synthesized by partial substitution of its carboxyl functionalities with furan, which could come into Diels-Alder click reaction with maleimide end groups of a four arm poly(ethylene glycol) cross-linker. Tuning the cooperative viscoelastic action of transient ionic and permanent click cross-links within the single network of alginate provided a soft tough hydrogel with a set of interesting features: (i) immediate self-recovery under cyclic loading, (ii) highly efficient and autonomous self-healing upon fracture, (iii) in situ forming ability for molding and minimally invasive injection, (iv) capability for viable cell encapsulation, and (v) reactivity for on-demand biomolecule conjugation. The facile strategy is applicable to a wide range of natural and synthetic polymers by introducing the calcium binding and click reacting functional groups and can broaden the use of tough hydrogels in load-bearing, cell-laden applications such as soft tissue engineering and bioactuators.


Subject(s)
Alginates/chemistry , Hydrogels/chemical synthesis , Cells, Cultured , Click Chemistry , Compressive Strength , Cross-Linking Reagents/chemistry , Elasticity , Humans , Hydrogels/pharmacology , Maleimides/chemistry , Myoblasts, Cardiac/drug effects , Polyethylene Glycols/chemistry , Viscosity
20.
J Biochem Mol Toxicol ; 32(11): e22213, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30152906

ABSTRACT

We investigated the effect of apigenin, a dietary flavonoid, on isoproterenol hydrochloride (ISO)-induced apoptotic signaling in cardiomyoblast H9C2 cells. The results showed that apigenin treatment (10 µM) prevented ISO (31.25 µM)-induced lipid peroxidative levels and antioxidants status in H9C2 cells. Furthermore, apigenin inhibited expression of inflammatory markers in ISO-treated cells. In addition, apigenin prevented ISO-induced DNA damage and apoptotic signaling through modulating the expression of Bax, caspase-3, -8 and -9, cytochrome c, and Fas proteins in H9C2 cells. It is concluded that apigenin prevents ISO-induced antioxidants depletion, oxidative DNA damage, inflammatory, and apoptotic signaling in H9C2 cells. Thus, the present results demonstrated that apigenin has a cardioprotective effect on cardiomyoblasts cells.


Subject(s)
Antioxidants/pharmacology , Apigenin/pharmacology , Apoptosis/drug effects , Cardiotonic Agents/adverse effects , Isoproterenol/adverse effects , Myoblasts, Cardiac/drug effects , Oxidative Stress/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Biomarkers/metabolism , Cardiotonic Agents/antagonists & inhibitors , Cell Line , DNA Damage/drug effects , Inflammation Mediators/agonists , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Isoproterenol/antagonists & inhibitors , Lipid Peroxidation/drug effects , Membrane Potential, Mitochondrial , Mitochondria, Heart/drug effects , Mitochondria, Heart/immunology , Mitochondria, Heart/metabolism , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/immunology , Mitochondrial Membranes/metabolism , Myoblasts, Cardiac/cytology , Myoblasts, Cardiac/immunology , Myoblasts, Cardiac/metabolism , Rats , bcl-2-Associated X Protein/agonists , bcl-2-Associated X Protein/antagonists & inhibitors , bcl-2-Associated X Protein/metabolism , fas Receptor/agonists , fas Receptor/antagonists & inhibitors , fas Receptor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL