Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Environ Toxicol ; 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39109685

ABSTRACT

Oxidative stress is a pivotal factor in the pathogenesis of various cardiovascular diseases. Rhodiola, a traditional Chinese medicine, is recognized for its potent antioxidant properties. Salidroside, a phenylpropanoid glycoside derived from Rhodiola rosea, has shown remarkable antioxidant capabilities. This study aimed to elucidate the potential protective mechanisms of Rhodiola and salidroside against H2O2-induced cardiac apoptosis in H9c2 cardiomyoblast cells. H9c2 cells were exposed to H2O2 for 4 h, and subsequently treated with Rhodiola or salidroside for 24 h. Cell viability and apoptotic pathways were assessed. The involvement of insulin-like growth factor 1 receptor (IGF1R) and the activation of extracellular regulated protein kinases 1/2 (ERK1/2) were investigated. H2O2 (100 µM) exposure significantly induced cardiac apoptosis in H9c2 cells. However, treatment with Rhodiola (12.5, 25, and 50 µg/mL) and salidroside (0.1, 1, and 10 nM) effectively attenuated H2O2-induced cytotoxicity and apoptosis. This protective effect was associated with IGF1R-activated phosphorylation of ERK1/2, leading to the inhibition of Fas-dependent proteins, HIF-1α, Bax, and Bak expression in H9c2 cells. The images from hematoxylin and eosin staining and immunofluorescence assays also revealed the protective effects of Rhodiola and salidroside in H9c2 cells against oxidative damage. Our findings suggest that Rhodiola and salidroside possess antioxidative properties that mitigate H2O2-induced apoptosis in H9c2 cells. The protective mechanisms involve the activation of IGF1R and subsequent phosphorylation of ERK1/2. These results propose Rhodiola and salidroside as potential therapeutic agents for cardiomyocyte cytotoxicity and apoptosis induced by oxidative stress in heart diseases. Future studies may explore their clinical applications in cardiac health.

2.
Drug Chem Toxicol ; 46(5): 1044-1050, 2023 Nov.
Article in English | MEDLINE | ID: mdl-36216784

ABSTRACT

To date, few studies have investigated the toxicological effects of the combined use of amphetamine and heroin in the heart. Hence, the aim of this study was to identify indicators for clinical evaluation and prevention of cardiac injury induced by the combined use of amphetamine and heroin. Four different groups were analyzed: (1) normal group (n=25;average age=35 ± 6.8); (2) heart disease group (n=25;average age=58 ± 17.2); (3) drug abusers (n = 27; average age = 37 ± 7.7); (4) drug abstainers (previous amphetamine-heroin users who had been drug-free for more than two weeks; n = 22; average age = 35 ± 5.6). The activity of MMPs, and levels of TNF-α, IL-6, GH, IGF-I, and several serum biomarkers were examined to evaluate the impact of drug abuse on the heart. The selected plasma biomarkers and classic cardiac biomarkers were significantly increased compared to the normal group. The zymography data showed the changes in cardiac-remodeling enzymes MMP-9 and MMP-2 among combined users of amphetamine and heroin. The levels of TNF-α and IL-6 only increased in the heart disease group. Growth hormone was increased; however, IGF-I level decreased with drug abuse and the level was not restored by abstinence. We speculated that the amphetamine-heroin users might pose risk to initiate heart disease even though the users abstained for more than two weeks. The activity change of MMP-9 and MMP-2 can be a direct reason affecting heart function. The indirect reason may be related to liver damage by drug abuse reduce IGF-1 production to protect heart function.


Subject(s)
Heart Diseases , Heart Injuries , Heroin Dependence , Humans , Adult , Middle Aged , Aged , Insulin-Like Growth Factor I , Matrix Metalloproteinase 2 , Matrix Metalloproteinase 9 , Heroin , Heroin Dependence/complications , Interleukin-6 , Tumor Necrosis Factor-alpha , Amphetamine , Biomarkers
3.
Acta Cardiol Sin ; 39(5): 687-694, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37720406

ABSTRACT

Background: Premature ventricular complex (PVC) without structural heart disease is mostly viewed as a benign arrhythmia. However, the high burden of PVC causes cardiomyopathy due to intraventricular dyssynchrony. The effects of ectopic contraction on left ventricular (LV) hemodynamics in the structurally normal heart are unclear. Objectives: To examine the effect of PVC burden on LV dimension, LV systolic function, and intraventricular blood flow, and to determine whether ectopic ventricular contraction affects LV hemodynamics. Methods: Patients aged ≥ 18 years with PVC ≥ 5% on Holter recording were enrolled and divided into groups G1 (5-10%), G2 (10-20%), and G3 (≥ 20%). We excluded patients with structural heart diseases, pacemakers, and LV systolic dysfunction [LV ejection fraction (LVEF) < 50%]. Clinical characteristics and routine transthoracic echocardiography parameters were compared. Results: The end-systolic LV internal dimension increased according to the PVC burden from G1 to G3 (p = 0.001). LVEF was inversely associated with PVC burden from G1 to G3 (p = 0.002). The same pattern was seen for LV outflow tract (LVOT) maximal velocity (p = 0.005) and maximal pressure gradient (PG) (p = 0.005), LVOT velocity time integral (VTI) (p = 0.03) and LV stroke volume index (LVSI) (p = 0.008). Conclusions: Systolic function and LV end-systolic dimension were inversely associated with PVC burden. Decreased LVOT flow velocity and PG were related to increased PVC burden. LVOT VTI and LVSI were smaller when the PVC burden exceeded 20%. These negative hemodynamic manifestations of idiopathic PVC were considerable even in structure normal hearts, hence the early elimination of PVC is strongly advised.

4.
Mol Cell Biochem ; 477(1): 143-152, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34586566

ABSTRACT

The heart is a very dynamic pumping organ working perpetually to maintain a constant blood supply to the whole body to transport oxygen and nutrients. Unfortunately, it is also subjected to various stresses based on physiological or pathological conditions, particularly more vulnerable to damages caused by oxidative stress. In this study, we investigate the molecular mechanism and contribution of IGF-IIRα in endoplasmic reticulum stress induction in the heart under doxorubicin-induced cardiotoxicity. Using in vitro H9c2 cells, in vivo transgenic rat cardiac tissues, siRNAs against CHOP, chemical ER chaperone PBA, and western blot experiments, we found that IGF-IIRα overexpression enhanced ER stress markers ATF4, ATF6, IRE1α, and PERK which were further aggravated by DOX treatment. This was accompanied by a significant perturbation in stress-associated MAPKs such as p38 and JNK. Interestingly, PARKIN, a stress responsive cellular protective mediator was significantly downregulated by IGF-IIRα concomitant with decreased expression of ER chaperone GRP78. Furthermore, ER stress-associated pro-apoptotic factor CHOP was increased considerably in a dose-dependent manner followed by elevated c-caspase-12 and c-caspase-3 activities. Conversely, treatment of H9c2 cells with chemical ER chaperone PBA or siRNA against CHOP abolished the IGF-IIRα-induced ER stress responses. Altogether, these findings suggested that IGF-IIRα contributes to ER stress induction and inhibits cellular stress coping proteins while increasing pro-apoptotic factors feeding into a cardio myocyte damage program that eventually paves the way to heart failure.


Subject(s)
Endoplasmic Reticulum Stress , Endoplasmic Reticulum/metabolism , Myocardium/metabolism , Receptor, IGF Type 2/metabolism , Animals , Cell Line , Cytotoxins/adverse effects , Cytotoxins/pharmacology , Doxorubicin/adverse effects , Doxorubicin/pharmacology , Endoplasmic Reticulum/genetics , Rats , Rats, Transgenic , Receptor, IGF Type 2/genetics
5.
Sleep Breath ; 26(3): 1161-1172, 2022 09.
Article in English | MEDLINE | ID: mdl-34626328

ABSTRACT

BACKGROUND: The purpose of this study was to investigate whether or not angiotensin II type 1 receptor blocker irbesartan (ARB) with a partial agonist of PPAR-γ could protect against chronic nocturnal intermittent hypoxia (CIH)-induced cardiac Fas/FasL-mediated to mitochondria-mediated apoptosis. METHODS: Sprague-Dawley rats were in a normoxic control group (CON-G), or rats were in a chronic nocturnal intermittent hypoxia group (HP-G, from 3 to 7% oxygen versus 21% oxygen per forty seconds cycle, nocturnally 8 h per day for 1 month), or rats were in a chronic nocturnal intermittent hypoxia group pretreated with ARB (50 mg/kg/day, S.C.) (ARB-HP-G). Echocardiography, H&E staining, TUNEL staining, and Western blotting were measured in the left ventricle. RESULTS: Hypoxia-induced SIRT1 degradation, Fas receptors, FADD, active caspase-8 and caspase-3 (Fas/FasL apoptotic pathway) and Bax, tBid, active caspase-9 and -3 (mitochondrial apoptotic pathway) and TUNEL-positive apoptosis were reduced in ARB-HP-G when compared with HP-G. IGF-I, IGF1 receptor, p-PI3k, p-Akt, Bcl2, and Bcl-XL (IGF1/PI3K/AKT pro-survival pathway) were increased in ARB-HP-G compared to HP-G. CONCLUSIONS: Our findings suggest that the ARB may prevent cardiac Fas/FasL to mitochondrial apoptotic pathways and enhance cardiac IGF1/PI3K/AKT pro-survival pathway in the sleep apnea model associated with JNK de-activation and SIRT1 upregulation. ARB prevents chronic sleep apnea-enhanced cardiac apoptosis via enhancing survival pathways.


Subject(s)
Sirtuin 1 , Sleep Apnea Syndromes , Angiotensin Receptor Antagonists , Angiotensin-Converting Enzyme Inhibitors , Animals , Apoptosis , Hypoxia , Irbesartan , Myocardium , Oxygen , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Rats , Rats, Sprague-Dawley , Up-Regulation
6.
Environ Toxicol ; 35(4): 468-477, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31794124

ABSTRACT

It has been well-documented that the consumption of deep sea water (DSW) has beneficial effects on myocardial hypertrophy and cardiac apoptosis induced by hypercholesterolemia. However, the molecular mechanisms for the anti-inflammatory effects of DSW on diabetic cardiomyopathy are still largely unclear. The main purpose of this present study was to test the hypothesis that DSW exerts anti-inflammatory effects through the suppression of the TNF-α-mediated signaling pathways. IP injection of streptozotocin (STZ) at the dose of 65 mg/kg was used to establish a diabetes rat model. DSW mineral extracts that diluted in desalinated water were prepared in three different dosages and administered to the rats through gavages for 4 weeks. These dosages are DSW-1X (equivalent to 37 mg Mg2+ /kg/day), 2X (equivalent to 74 mg Mg2+ /kg/day) and 3X (equivalent to 111 mg Mg2+ mg/kg/day). Immunofluorescence staining and Western blot showed that the protein expression level of TNF-α was markedly higher in the STZ-induced diabetic rat hearts than in the control group. Consequently, the phosphorylation levels of the TNF-α-modulated downstream signaling molecules and P38 mitogen-activated protein kinases (MAPKs) were notably elevated in heart tissues of STZ-induced diabetes. These higher phosphorylation levels subsequently upregulated NF-κB-modulated inflammatory mediators, such as cyclooxygenase (COX)-II and inducible nitric oxide synthase (iNOS). However, treatment with DSW as well as MgSO4 , the main mineral in DSW, significantly reversed all the alterations. These findings suggest that DSW has potential as a therapeutic agent for preventing diabetes-related cardiovascular diseases.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Diabetic Cardiomyopathies/prevention & control , Minerals/therapeutic use , Seawater/chemistry , Tumor Necrosis Factor-alpha/metabolism , Animals , Anti-Inflammatory Agents/administration & dosage , Diabetes Mellitus, Experimental/immunology , Diabetic Cardiomyopathies/immunology , Inflammation , Male , Minerals/administration & dosage , Myocardium/immunology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/immunology , Myocytes, Cardiac/pathology , Rats , Rats, Sprague-Dawley , Signal Transduction , Streptozocin
7.
Am J Physiol Cell Physiol ; 317(2): C235-C243, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31116582

ABSTRACT

Doxorubicin (DOX) is an anthracycline antibiotic commonly employed for the treatment of various cancers. However, its therapeutic uses are hampered by side effects associated with cumulative doses during the course of treatment. Whereas deregulation of autophagy in the myocardium has been involved in a variety of cardiovascular diseases, the role of autophagy in DOX-induced cardiomyopathy remains debated. Our earlier studies have shown that DOX treatment in a rat animal model leads to increased expression of the novel stress-inducible protein insulin-like growth factor II receptor-α (IGF-IIRα) in cardiac tissues, which exacerbated the cardiac injury by enhancing oxidative stress and p53-mediated mitochondria-dependent cardiac apoptosis. Through this study, we investigated the contribution of IGF-IIRα to dysregulation of autophagy in heart using both in vitro H9c2 cells (DOX treated, 1 µM) and in vivo transgenic rat models (DOX treated, 5 mg/kg ip for 6 wk) overexpressing IGF-IIRα specifically in the heart. We found that IGF-IIRα primarily localized to mitochondria, causing increased mitochondrial oxidative stress that was severely aggravated by DOX treatment. This was accompanied by a significant perturbation in mitochondrial membrane potential and increased leakage of cytochrome c, causing increased cleaved caspase-3 activity. There were significant alterations in phosphorylated AMP-activated protein kinase (p-AMPK), phosphorylated Unc-51 like kinase-1 (p-ULK1), PARKIN, PTEN-induced kinase 1 (PINK1), microtubule-associated protein 1 light chain 3 (LC3), and p62 proteins, which were more severely disrupted under the combined effect of IGF-IIRα overexpression plus DOX. Finally, LysoTracker Red staining showed that IGF-IIRα overexpression causes lysosomal impairment, which was rescued by rapamycin treatment. Taken together, we found that IGF-IIRα leads to mitochondrial oxidative stress, decreased antioxidant levels, disrupted mitochondrial membrane potential, and perturbed mitochondrial autophagy contributing to DOX-induced cardiomyopathy.


Subject(s)
Antibiotics, Antineoplastic/toxicity , Doxorubicin/toxicity , Heart Diseases/chemically induced , Mitochondria, Heart/drug effects , Mitophagy/drug effects , Myocytes, Cardiac/drug effects , Oxidative Stress/drug effects , Receptor, IGF Type 2/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Autophagy-Related Proteins/metabolism , Cardiotoxicity , Cell Line , Heart Diseases/genetics , Heart Diseases/metabolism , Heart Diseases/pathology , Lysosomes/drug effects , Lysosomes/metabolism , Lysosomes/pathology , Membrane Potential, Mitochondrial/drug effects , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rats, Sprague-Dawley , Rats, Transgenic , Receptor, IGF Type 2/genetics , Signal Transduction/drug effects
8.
J Cell Biochem ; 120(10): 16956-16966, 2019 10.
Article in English | MEDLINE | ID: mdl-31104312

ABSTRACT

Cardiotoxicity by doxorubicin hampers its therapeutic potential as an anticancer drug, but mechanisms leading to cardiotoxicity remain contentious. Through this study, the functional contribution of insulin-like growth factor receptor type II α (IGF-IIRα) which is a novel stress-inducible protein was explored in doxorubicin-induced cardiac stress. Employing both in vitro H9c2 cells and in vivo transgenic rat models (SD-TG [IGF-IIRα]) overexpressing IGF-IIRα specifically in heart, we found that IGF-IIRα leads to cardiac structural abnormalities and functional perturbations that were severely aggravated by doxorubicin-induced cardiac stress. Overexpression of IGF-IIRα leads to cumulative elevation of stress associated cardiac hypertrophy and apoptosis factors. There was a significant reduction of survival associated proteins p-Akt and estrogen receptor ß/α, and abnormal elevation of cardiac hypertrophy markers such as atrial natriuretic peptide, cardiac troponin-I, and apoptosis-inducing agents such as p53, Bax, and cytochrome C, respectively. IGF-IIRα also altered the expressions of AT1R, ERK1/2, and p38 proteins. Besides, IGF-IIRα also increased the reactive oxygen species production in H9c2 cells which were markedly aggravated by doxorubicin treatment. Together, we showed that IGF-IIRα is a novel stress-induced protein that perturbed cardiac homeostasis and cumulatively exacerbated the doxorubicin-induced cardiac injury that perturbed heart functions and ensuing cardiomyopathy.


Subject(s)
Antibiotics, Antineoplastic/toxicity , Cardiomegaly/chemically induced , Cardiomyopathies/chemically induced , Doxorubicin/toxicity , Heart Defects, Congenital/chemically induced , Receptor, IGF Type 2/biosynthesis , Animals , Apoptosis/drug effects , Cardiotoxicity/pathology , Cell Line , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Heart/anatomy & histology , Myocytes, Cardiac/metabolism , Oxidative Stress/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Transgenic , Reactive Oxygen Species/metabolism , Receptor, IGF Type 2/genetics , Signal Transduction/drug effects
9.
Environ Toxicol ; 34(2): 210-218, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30450786

ABSTRACT

IGF-IIR activation regulates cardiac remodeling leading to apoptosis. Here, we identified the novel IGF-IIRα (150 KDa), a truncated IGF-IIR transcript enhances cardiac apoptosis under high-salt uptake in transgenic rat model. Echocardiographic analysis revealed decline in ejection fraction and fractional shortening percentage in IGF-IIRα (TG) rats. We found that IGF-IIRα TG rats developed severe apoptosis and fibrosis as identified through TUNEL assay and Masson's trichrome staining. Importantly, the heart functioning, apoptosis, and fibrosis were significantly affected under high-salt conditions in IGF-IIRα (TG) rats. Significant upregulation of apoptosis was evident from decreased Bcl-2, p-AKT, and p-PI3K expressions with concomitant increase in Bad, cytochrome C, cleaved caspase 3 levels. We found that, IGF-IIRα highly induced tissue fibrosis through collagen accumulation (col I, col III) and up regulated various fibrotic markers such as tPA, uPA, TGF-ß, and vimentin expressions. The observed upregulation of fibrosis were significantly regulated under high-salt conditions and their over regulation under IGF-IIRα over expressions shows the key role of IGF-IIRα in promoting high-salt induced fibrosis. During IGF-IIRα over expression induced cardiotoxicity, under high salt condition, and it destroys the interaction between CHIP and HSF1, which promotes the degradation of HSF1 and results in upregulation of IGF-IIR/IGF-IIRα expressions. Altogether, the study unveils novel IGF-IIRα in the regulation of cardiac apoptosis and fibrosis under high-salt diet.


Subject(s)
Apoptosis/genetics , Gene Expression Regulation , Myocardium/pathology , Receptor, IGF Type 2/genetics , Sodium Chloride, Dietary/adverse effects , Ventricular Remodeling/genetics , Animals , Apoptosis/drug effects , Fibrosis , Gene Expression Regulation/drug effects , Myocardium/metabolism , Rats , Rats, Transgenic , Ventricular Remodeling/drug effects
10.
Int J Mol Sci ; 20(17)2019 Sep 02.
Article in English | MEDLINE | ID: mdl-31480672

ABSTRACT

Cardiovascular diseases have a high prevalence worldwide and constitute the leading causes of mortality. Recently, malfunctioning of ß-catenin signaling has been addressed in hypertensive heart condition. Ang-II is an important mediator of cardiovascular remodeling processes which not only regulates blood pressure but also leads to pathological cardiac changes. However, the contribution of Ang-II/ß-catenin axis in hypertrophied hearts is ill-defined. Employing in vitro H9c2 cells and in vivo spontaneously hypertensive rats (SHR) cardiac tissue samples, western blot analysis, luciferase assays, nuclear-cytosolic protein extracts, and immunoprecipitation assays, we found that under hypertensive condition ß-catenin gets abnormally induced that co-activated LEF1 and lead to cardiac hypertrophy changes by up-regulating the IGF-IIR signaling pathway. We identified putative LEF1 consensus binding site on IGF-IIR promoter that could be regulated by ß-catenin/LEF1 which in turn modulate the expression of cardiac hypertrophy agents. This study suggested that suppression of ß-catenin expression under hypertensive condition could be exploited as a clinical strategy for cardiac pathological remodeling processes.


Subject(s)
Cardiomegaly/chemically induced , Cardiomegaly/metabolism , Receptor, IGF Type 2/metabolism , Signal Transduction , beta Catenin/metabolism , Angiotensin II , Animals , Biomarkers/metabolism , Cardiomegaly/pathology , Cell Nucleus/metabolism , GATA4 Transcription Factor/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Lymphoid Enhancer-Binding Factor 1/metabolism , Myocytes, Cardiac/metabolism , NFATC Transcription Factors/metabolism , Promoter Regions, Genetic/genetics , Protein Kinase C-alpha/metabolism , Rats, Inbred SHR
11.
J Mol Cell Cardiol ; 122: 58-68, 2018 09.
Article in English | MEDLINE | ID: mdl-30098987

ABSTRACT

Mitochondrial dysfunction is a major contributor to myocyte loss and the development of heart failure. Myocytes have quality control mechanisms to retain functional mitochondria by removing damaged mitochondria via specialized autophagy, i.e., mitophagy. The underlying mechanisms of fission affect the survival of cardiomyocytes, and left ventricular function in the heart is poorly understood. Here, we demonstrated the direct effect and potential mechanisms of mitochondrial functional defects associated with abnormal mitochondrial dynamics in heart failure. We observed that IGF-IIR signaling produced significant changes in mitochondrial morphology and function; such changes were associated with the altered expression and distribution of dynamin-related protein (Drp1) and mitofusin (Mfn2). IGF-IIR signaled extracellular signal-regulated kinase (ERK) activation to promote Drp1 phosphorylation and translocation to mitochondria for mitochondrial fission and mitochondrial dysfunction. Moreover, IGF-IIR signaling triggered Rab9-dependent autophagosome formation by the JNK-mediated phosphorylation of Bcl-2 at serine 87 and promoted ULK1/Beclin 1-dependent autophagic membrane formation. Excessive mitochondrial fission by Drp1 enhanced the Rab9-dependent autophagosome recognition and engulfing of damaged mitochondria and eventually decreased cardiomyocyte viability. Therefore, these results demonstrated the connection between Rab9-dependent autophagosomes and mitochondrial fission in cardiac myocytes, which provides a potential therapeutic strategy for treating heart disease.


Subject(s)
Dynamins/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Heart Failure/metabolism , Mitochondria, Heart/metabolism , Receptor, IGF Type 2/metabolism , Analysis of Variance , Animals , Autophagosomes/metabolism , Autophagy , Cell Line , Female , MAP Kinase Signaling System , Mitochondrial Dynamics , Mitophagy , Myocytes, Cardiac/metabolism , Phosphorylation , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Sprague-Dawley , rab GTP-Binding Proteins/metabolism
12.
J Cell Physiol ; 233(1): 463-475, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28295305

ABSTRACT

Doxorubicin (DOX), one useful chemotherapeutic agent, is limited in clinical use because of its serious cardiotoxicity. Growing evidence suggests that angiotensin receptor blockers (ARBs) have cardioprotective effects in DOX-induced cardiomyopathy. However, the detailed mechanisms underlying the action of ARBs on the prevention of DOX-induced cardiomyocyte cell death have yet to be investigated. Our results showed that angiotensin II receptor type I (AT1 R) plays a critical role in DOX-induced cardiomyocyte apoptosis. We found that MAPK signaling pathways, especially ERK1/2, participated in modulating AT1 R gene expression through DOX-induced mitochondrial ROS release. These results showed that several potential heat shock binding elements (HSE), which can be recognized by heat shock factors (HSFs), located at the AT1 R promoter region. HSF2 markedly translocated from the cytoplasm to the nucleus when cardiomyocytes were damaged by DOX. Furthermore, the DNA binding activity of HSF2 was enhanced by DOX via deSUMOylation. Overexpression of HSF2 enhanced DOX-induced cardiomyocyte cell death as well. Taken together, we found that DOX induced mitochondrial ROS release to activate ERK-mediated HSF2 nuclear translocation and AT1 R upregulation causing DOX-damaged heart failure in vitro and in vivo.


Subject(s)
Antibiotics, Antineoplastic , Doxorubicin , Heart Diseases/enzymology , Heat-Shock Proteins/metabolism , Mitochondria, Heart/enzymology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Myocytes, Cardiac/enzymology , Reactive Oxygen Species/metabolism , Receptor, Angiotensin, Type 1/metabolism , Transcription Factors/metabolism , Active Transport, Cell Nucleus , Animals , Apoptosis , Binding Sites , Cardiotoxicity , Cell Line , Disease Models, Animal , Enzyme Activation , Heart Diseases/chemically induced , Heart Diseases/genetics , Heart Diseases/pathology , Mitochondria, Heart/pathology , Myocytes, Cardiac/pathology , Promoter Regions, Genetic , RNA Interference , Rats, Inbred WKY , Receptor, Angiotensin, Type 1/genetics , Signal Transduction , Sumoylation , Transfection , Up-Regulation
13.
J Cell Physiol ; 233(9): 7080-7091, 2018 09.
Article in English | MEDLINE | ID: mdl-29574782

ABSTRACT

Mitochondria dysfunction is the major characteristic of mitophagy, which is essential in mitochondrial quality control. However, excessive mitophagy contributes to cell death in a number of diseases, including ischemic stroke and hepatotoxicity. Insulin-like growth factor II (IGF-II) and its receptor (IGF-IIR) play vital roles in the development of heart failure during hypertension. We found that IGF-II triggers IGF-IIR receptor activation, causing mitochondria dysfunction, resulting in mitophagy, and cardiomyocyte cell death. These results indicated that IGF-IIR activation triggers mitochondria fragmentation, leading to autophagosome formation, and loss of mitochondria content. These results are associated with Parkin-dependent mitophagy. Additionally, autophagic proteins Atg5, and Atg7 deficiency did not suppress IGF-IIR-induced mitophagy. However, Rab9 knockdown reduced mitophagy and maintained mitochondrial function. These constitutive mitophagies through IGF-IIR activation trigger mitochondria loss and mitochondrial ROS accumulation for cardiomyocyte viability decrease. Together, our results indicate that IGF-IIR predominantly induces mitophagy through the Rab9-dependent alternative autophagy.


Subject(s)
Autophagy , Mitochondria/metabolism , Mitophagy , Receptor, IGF Type 2/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Animals, Newborn , Autocrine Communication , Autophagosomes/metabolism , Autophagosomes/ultrastructure , Dependovirus/metabolism , Female , Heart/physiopathology , Humans , Insulin-Like Growth Factor II/metabolism , Mitochondria/ultrastructure , Models, Biological , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/ultrastructure , Organ Specificity , Paracrine Communication , Rats, Sprague-Dawley , Ubiquitin-Protein Ligases/metabolism
14.
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
15.
Environ Toxicol ; 33(1): 93-103, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29087013

ABSTRACT

BACKGROUND: Doxorubicin (Dox) is an effective anticancer agent. However, its effectiveness is limited by its cardiotoxic effects. It has also been reported that the mitogen-activated protein kinase family and NF-κB can be activated by Dox treatment. DATS has been shown to be a potent antioxidant with cardioprotective effects. We investigate whether Dox induces cardiac apoptosis through JNK- and ERK-dependent NF-κB upregulation that can be reduced by DATS treatment. METHODS AND MATERIAL: H9c2 cells were treated with 0.5-1.5 µM Dox for 24 hours. Dox promoted apoptosis and ROS generation and inhibited viability in a dose-dependent manner. Then, the phosphorylation levels of JNK, ERK, and NF-κB evaluated by western blot were elevated. We used inhibitors of JNK, ERK, and NF-κB to determine which of these proteins were involved in Dox-induced apoptosis. Furthermore, Dox-exposed cells were treated with DATS at doses of 1, 5, and 10 µM, and the data demonstrated that ROS generation and apoptotic proteins were decreased and that ERK and NF-κB were downregulated in a dose-dependent manner. Additionally, six-week-old rats were divided into three groups (n = 6 per group) designed as an eight-week study. Normal, Dox (at dose 3.75 mg/kg by ip) administered with or without DATS (at dose 40 mg/kg by gavage) treatment groups. The results indicate that cardiac dysfunction, apoptosis, and JNK, ERK, and NF-κB activation by Dox were reversed by treatment with DATS. CONCLUSION: DATS appears to suppress Dox-induced cardiomyocyte apoptosis by inhibiting NADPH oxidase-related ROS production and the downstream JNK/ERK/NF-κB signaling pathway; DATS may possess clinical therapeutic potential by blocking Dox-induced cardiotoxicity.


Subject(s)
Allyl Compounds/pharmacology , Apoptosis/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Sulfides/pharmacology , Acetylcysteine/pharmacology , Animals , Apoptosis Regulatory Proteins/metabolism , Cell Line , Doxorubicin/toxicity , Extracellular Signal-Regulated MAP Kinases/metabolism , Heart/drug effects , JNK Mitogen-Activated Protein Kinases/metabolism , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , NF-kappa B/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar
16.
J Recept Signal Transduct Res ; 37(5): 493-499, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28812967

ABSTRACT

Cardiomyopathy involves changes in myocardial ultrastructure and cardiac hypertrophy. Angiotensin II (AngII) has previously been shown to stimulate the expression of IGF-2 and IGF-2R in H9c2 cardiomyoblasts and increase of blood pressure, and cardiac hypertrophy. Estrogen receptors (ERs) exert protective effects, such as anti-hypertrophy in cadiomyocytes. Tanshinone IIA (TSN), a main active ingredient from a Chinese medical herb, Salvia miltiorrhiza Bunge (Danshen), was shown to protect cardiomyocytes hypertrophy by different stress signals. We aimed to investigate whether TSN protected H9c2 cardiomyocytes from AngII-induced activation of IGF-2R pathway and hypertrophy by mediating through ERs. AngII resulted in H9c2 cardiomyoblast hypertrophy and increased inflammatory molecular markers. These were down-regulated by TSN via estrogen receptors. AngII resulted in elevation in MAPKs, IGF-2R and hypertrophic protein markers. These, again, were reduced by addition of the phytoestrogen with activation of ERs. Finally, AngII induced phosphorylation of heat shock factor-1 (HSF1) and decreased sirtuin-1 (SIRT1). In addition, AngII also caused an increase in distribution of IGF-2R molecules on cell membrane. In contrast, TSN reduced HSF1 phosphorylation and cell surface IGF-2R while elevating SIRT1 via ERs. TSN was capable of attenuating AngII-induced IGF-2R pathway and hypertrophy through ERs in H9c2 cardiomyoblast cells.


Subject(s)
Abietanes/administration & dosage , Cardiomegaly/drug therapy , Insulin-Like Growth Factor II/genetics , Receptor, IGF Type 2/genetics , Angiotensin II/administration & dosage , Angiotensin II/metabolism , Animals , Cardiomegaly/genetics , Cardiomegaly/pathology , Cell Line , Drugs, Chinese Herbal/administration & dosage , Gene Expression/drug effects , Heat Shock Transcription Factors/genetics , Humans , Insulin-Like Growth Factor II/metabolism , Mitogen-Activated Protein Kinase Kinases/genetics , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rats , Receptor, IGF Type 2/metabolism , Receptors, Estrogen/genetics , Signal Transduction/drug effects , Sirtuin 1/genetics
17.
Mol Cell Biochem ; 434(1-2): 181-195, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28577190

ABSTRACT

The present study was aimed to investigate the protective effects of 17ß-estradiol (E2) and estrogen receptor α (ERα) on isoproterenol (ISO)-treated H9c2 cardiomyoblast cells. In the present study, we treated H9c2 cells with ISO, a ß-adrenergic receptor agonist, to induce myocardiac hypertrophy. Pre-administration of E2 or ERα (induced by doxycycline) and E2 plus ERα significantly prevented ISO-induced increase of cell size and cytosolic calcium accumulation, accompanied with increased mRNA of atrial natriuretic peptide and brain natriuretic peptide. However, ICI-ERs antagonist, and melatonin, a specific inhibitor for ERα, reversed the cardioprotective effects, suggesting that E2 action was mediated through ERα. Further evidences showed that E2 and ERα increased the protein level of GSK3ß and protein phosphatase 2a inhibitor 2 (I2-PP2A), which subsequently enhanced the activation of I2-PP2A by disrupting PP2A activity and maintains normal calcium outflow. Collectively, E2 and ERα inhibited hypertrophy by preventing cytosol calcium accumulation and by inhibiting the association between PP2A with Na+-Ca2+ exchanger via GSK3ß and I2-PP2A activation.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Atrial Natriuretic Factor/metabolism , Calcium/metabolism , Estradiol/physiology , Estrogen Receptor alpha/physiology , Glycogen Synthase Kinase 3 beta/metabolism , Isoproterenol/pharmacology , NFATC Transcription Factors/metabolism , Protein Phosphatase 2/metabolism , Animals , Cell Line
18.
Environ Toxicol ; 32(2): 679-689, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27087047

ABSTRACT

Q10 is a powerful antioxidant often used in medical nutritional supplements for cancer treatment. This study determined whether Q10 could effectively prevent cardio-toxicity caused by doxorubicin treatment. Four week old SD rats were segregated into groups namely control, doxorubicin group (challenged with doxorubicin), Dox + Q10 group (with doxorubicin challenge and oral Q10 treatment), and Q10 group (with oral Q10 treatment). Doxorubicin groups received IP doxorubicin (2.5 mg/kg) every 3 days and Q10 groups received Q10 (10 mg/kg) every day. Three weeks of doxorubicin challenge caused significant reduction in heart weight, disarray in cardiomyocyte arrangement, elevation of collagen accumulation, enhancement of fibrosis and cell death associated proteins, and inhibition of survival proteins. However, Q10 effectively protected cardiomyocytes and ameliorated fibrosis and cell death induced by doxorubicin. Q10 is, therefore, evidently a potential drug to prevent heart damage caused by doxorubicin. © 2016 Wiley Periodicals, Inc. Environ Toxicol 32: 679-689, 2017.


Subject(s)
Antibiotics, Antineoplastic/adverse effects , Cardiomyopathies/prevention & control , Cardiotonic Agents/pharmacology , Doxorubicin/adverse effects , Ubiquinone/analogs & derivatives , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Cardiomyopathies/chemically induced , Cell Survival/drug effects , Male , Myocytes, Cardiac/drug effects , Rats , Rats, Sprague-Dawley , Ubiquinone/pharmacology
19.
Chin J Physiol ; 60(3): 166-173, 2017 Jun 30.
Article in English | MEDLINE | ID: mdl-28629210

ABSTRACT

In this study, a rat carotid balloon injury-animal model was used to elucidate the temporal relation of hypertrophy in the progression of cardiac damage and the role of insulin-like growth factor (IGF)-I survival pathway on course of the cardiac damage. Rats were subjected to carotid balloon-injury and examined at different time points. We further studied the heart-weight/body-weight-ratio, histology and protein expression to understand the pathological events associated with percutaneous transluminal coronary angioplasty (PTCA) induced damages. Protein expression analysis showed increased levels of IGF-I signaling pathway and mitogen-activated protein kinase (MAPK) signaling pathway after 2 h and after 2 d of carotid balloon injury. On the other hand, apoptosis signaling pathways were enhanced after 14 d of carotid balloon injury. According to the results, rat carotid balloon injury significantly induced IGF-I survival signaling and compensated hypertrophy pathway during the initial period of injury however after 14 d the proteins involved in apoptotic cell death were elevated and the proteins of the survival pathway and compensatory hypertrophy were significantly reduced.


Subject(s)
Apoptosis/physiology , Carotid Arteries/metabolism , Insulin-Like Growth Factor I/metabolism , Signal Transduction/physiology , Animals , Heart/physiology , Hypertrophy/metabolism , Male , Mitogen-Activated Protein Kinases/metabolism , Proteins/metabolism , Rats , Rats, Wistar
20.
Int J Mol Sci ; 18(3)2017 Mar 14.
Article in English | MEDLINE | ID: mdl-28335423

ABSTRACT

Estrogen receptor α (ERα) and estrogen receptor ß (ERß) play important roles in cardiovascular disease (CVD) prevention. Recently, these estrogen receptors were reconsidered as an important treatment target of obesity leading to CVD. In this study, 17ß-estradiol (17ß-E) replacement therapy applied to high-fat diet-induced obese C57B male mice and ovariectomized (OVX) rats were evaluated, and the protective effects against high-fat diet-induced obesity were assessed in C57B mouse hearts. The results showed that 17ß-E treatment activated both ERα and ERß, and ERß levels increased in a dose-dependent manner in high-fat diet C57B mouse cardiomyocytes following 17ß-E treatment. Notably, an almost 16% reduction in body weight was observed in the 17ß-E-treated (12 µg/kg/day for 60 days) high-fat diet-induced obese C57B male mice. These results suggested that 17ß-E supplements may reduce CVD risk due to obesity.


Subject(s)
Body Weight , Cardiovascular Diseases/prevention & control , Estradiol/therapeutic use , Estrogens/therapeutic use , Obesity/drug therapy , Animals , Cardiovascular Diseases/etiology , Diet, High-Fat/adverse effects , Female , Male , Mice , Mice, Inbred C57BL , Obesity/complications , Obesity/etiology , Obesity/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Estrogen/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL