Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 15 de 15
1.
Environ Toxicol ; 38(2): 436-450, 2023 Feb.
Article En | MEDLINE | ID: mdl-36421005

Thioredoxin reductase 3 (Txnrd3) plays a crucial role in antioxidant and anti-cancer activities, and sperm maturation. The damage of heavy metals, including Nickel (Ni), is the most prominent harm in social development, and hampering Txnrd3 might exacerbate Ni-induced cardiac damage. In this study, a total of 160 8-week-old C57BL/N male mice with 25-30 g weight of Txnrd3+/+ wild-type and Txnrd3-/- homozygote-type were randomly divided into eight groups. The mice in the control and Ni groups were gavaged with distilled water and a freshly prepared 10 mg/kg NiCl2 solution. Melatonin (Mel) groups were administered at a concentration of 2 mg/kg for 21 days at the mice's 0.1 ml/10 g body weight. Ni exposure up-regulated the messenger RNA (mRNA) levels of mitochondrial apoptosis (caspase-3, caspase-9, cytochrome c, p53, and BAX), autophagy (LC3, ATG 1, ATG 7, and Beclin-1), and inflammation (TNF-α, COX 2, IL-1ß, IL-2, IL-6, and IL-7)-related markers, but down-regulated the mRNA levels of BCL-2, p62 and mTOR (p < .05). Ni exposure decreased the expression of BCL-2 and p62 protein but increased the expression levels of caspase-3, caspase-9, cytochrome c, p53, BAX, ATG 7, Beclin-1, TNF-α, COX 2, IL-1ß and IL-2 protein (p < .05). Ni increased the contents of glutathione disulfide (GSSG) and malondialdehyde (MDA) and decreased the activities of catalase (CAT) and total superoxide dismutase (T-SOD) (p < .05). Decreased Txnrd3 expression significantly exacerbated changes compared to the Ni exposure (p < .05). Mel significantly attenuated these changes, but the effect decreased when Txnrd3 was inhibited (p < .05). In conclusion, decreased Txnrd3 expression promoted Ni-induced mitochondrial apoptosis and inflammation via oxidative stress and aggravated heart damage in mice. Decreased Txnrd3 expression significantly reduced the protective effect of Mel to Ni exposure.


Apoptosis , Cardiotoxicity , Interleukin-2 , Nickel , Oxidative Stress , Thioredoxin-Disulfide Reductase , Animals , Male , Mice , bcl-2-Associated X Protein/metabolism , Beclin-1/metabolism , Caspase 3/metabolism , Caspase 9/metabolism , Cyclooxygenase 2/metabolism , Cytochromes c/metabolism , Inflammation/chemically induced , Interleukin-2/metabolism , Mice, Inbred C57BL , Nickel/toxicity , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/metabolism , Thioredoxin-Disulfide Reductase/genetics , Thioredoxin-Disulfide Reductase/metabolism , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor Protein p53/metabolism , Cardiotoxicity/enzymology
2.
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
3.
Biomed Res Int ; 2020: 3189672, 2020.
Article En | MEDLINE | ID: mdl-33274202

Diabetes mellitus has developed into one of the debilitating diseases disturbing the health of many people living with cardiovascular diseases in modern times. The root of Ageratum conyzoides was investigated for its effects on alloxan-induced diabetic Wistar rats' cardiac tissues. Thirty-two (32) Wistar rats weighing between 180 and 190 g were randomly divided into four groups. The animals in groups B-D were induced with a single dose of 150 mg/kg body weight of alloxan (ALX) intraperitoneally. They were confirmed hyperglycemic after 72 hours of induction and then sustained in hyperglycemic condition for 2 weeks. Animals in groups C and D received AC intervention, as stated above, for four weeks. The body weight of the experimental animals and blood collection for glucose estimation were taken weekly for six weeks using appropriate instruments. Biochemical assays for lipid profile, antioxidant enzymatic, and nonenzymatic markers were carried out. Histopathological changes in the cardiac tissues were also studied. Administration of 150 mg/kg of ALX to experimental rats induced diabetes and significantly reduced the body weights, significantly (p < 0.05) increased the glucose level, triglyceride (TG), total cholesterol (TC), and low-density lipoprotein (LDL) levels, and decreased the levels of high-density lipoprotein (HDL) and antioxidant enzymatic markers such as catalase (CAT), superoxide dismutase (SOD), and glutathione peroxidase (GPx) while the antioxidant nonenzymatic marker such as malondialdehyde (MDA) level was significantly increased. By contrast, rats given the ethanolic extract root of A. conyzoides had significantly (p < 0.05) increased the body weight gain, whereas the glucose levels significantly (p < 0.05) improved in treated diabetic rats. This extract also improved the cardiovascular system of the diabetic rats by significantly decreasing TG and LDL levels, significantly (p < 0.05) increasing the HDL level, significantly reducing the cardiac contents of CAT, SOD, and GPx, and significantly (p < 0.05) decreasing MDA. Ethanolic extract root of A. conyzoides exhibited antihyperglycemic and antihyperlipidemic activities and mitigates damage to the heart from the ALX-induced myocardial toxicity associated with type-1 diabetes.


Ageratum/chemistry , Cardiotonic Agents/therapeutic use , Cardiotoxicity/drug therapy , Diabetes Mellitus, Experimental/drug therapy , Plant Extracts/therapeutic use , Plant Roots/chemistry , Administration, Oral , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Biomarkers/metabolism , Blood Glucose/metabolism , Body Weight/drug effects , Cardiotonic Agents/pharmacology , Cardiotoxicity/blood , Cardiotoxicity/enzymology , Cardiotoxicity/pathology , Diabetes Mellitus, Experimental/blood , Ethanol , Female , Lipids/blood , Male , Malondialdehyde/metabolism , Pancreas/drug effects , Pancreas/pathology , Plant Extracts/pharmacology , Rats, Wistar
4.
J Cell Mol Med ; 24(12): 6534-6557, 2020 06.
Article En | MEDLINE | ID: mdl-32336039

Anthracyclines is an effective chemotherapeutic treatment used for many types of cancer. However, high cumulative dosage of anthracyclines leads to cardiac toxicity and heart failure. Dysregulation of mitochondrial dynamics and function are major pathways driving this toxicity. Several pharmacological and non-pharmacological interventions aiming to attenuate cardiac toxicity by targeting mitochondrial dynamics and function have shown beneficial effects in cell and animal models. However, in clinical practice, there is currently no standard therapy for the prevention of anthracycline-induced cardiotoxicity. This review summarizes current reports on the impact of anthracyclines on cardiac mitochondrial dynamics and mitochondrial function and potential interventions targeting these pathways. The roles of mitochondrial dynamics and mitochondrial function in the development of anthracycline-induced cardiotoxicity should provide insights in devising novel strategies to attenuate the cardiac toxicity induced by anthracyclines.


Cardiotoxicity/metabolism , Cardiotoxicity/pathology , Doxorubicin/adverse effects , Mitochondria, Heart/pathology , Mitochondrial Dynamics , Adenylate Kinase/metabolism , Animals , Cardiotoxicity/enzymology , Humans , Iron/metabolism
5.
J Cell Mol Med ; 23(7): 4627-4639, 2019 07.
Article En | MEDLINE | ID: mdl-31106979

Doxorubicin (DOX) is widely used to treat various cancers affecting adults and children; however, its clinical application is limited by its cardiotoxicity. Previous studies have shown that children are more susceptible to the cardiotoxic effects of DOX than adults, which may be related to different maturity levels of cardiomyocyte, but the underlying mechanisms are not fully understood. Moreover, researchers investigating DOX-induced cardiotoxicity caused by human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have shown that dexrazoxane, the recognized cardioprotective drug for treating DOX-induced cardiotoxicity, does not alleviate the toxicity of DOX on hiPSC-CMs cultured for 30 days. We have suggested that this may be ascribed to the immaturity of the 30 days hiPSC-CMs. In this study, we investigated the mechanisms of DOX induced cardiotoxicity in cardiomyocytes of different maturity. We selected 30-day-old and 60-day-old hiPSC-CMs (day 30 and day 60 groups), which we term 'immature' and 'relatively mature' hiPSC-CMs, respectively. The day 30 CMs were found to be more susceptible to DOX than the day 60 CMs. DOX leads to more ROS (reactive oxygen species) production in the day 60 CMs than in the relatively immature group due to increased mitochondria number. Moreover, the day 60 CMs mainly expressed topoisomerase IIß presented less severe DNA damage, whereas the day 30 CMs dominantly expressed topoisomerase IIα exhibited much more severe DNA damage. These results suggest that immature cardiomyocytes are more sensitive to DOX as a result of a higher concentration of topoisomerase IIα, which leads to more DNA damage.


Cardiotoxicity/enzymology , Cardiotoxicity/pathology , Cell Differentiation , DNA Topoisomerases, Type II/metabolism , Doxorubicin/adverse effects , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/enzymology , Poly-ADP-Ribose Binding Proteins/metabolism , Cell Death/drug effects , Cells, Cultured , DNA Damage , Humans , Models, Biological , Reactive Oxygen Species/metabolism , Time Factors
6.
PLoS One ; 14(4): e0215992, 2019.
Article En | MEDLINE | ID: mdl-31034488

The clinical use of the chemotherapeutic doxorubicin (Dox) is limited by cardiotoxic side-effects. One of the early Dox effects is induction of a sarcoplasmic reticulum (SR) Ca2+ leak. The chaperone Glucose regulated protein 78 (GRP78) is important for Ca2+ homeostasis in the endoplasmic reticulum (ER)-the organelle corresponding to the SR in non-cardiomyocytes-and has been shown to convey resistance to Dox in certain tumors. Our aim was to investigate the effect of cardiac GRP78 gene transfer on Ca2+ dependent signaling, cell death, cardiac function and survival in clinically relevant in vitro and in vivo models for Dox cardiotoxicity.By using neonatal cardiomyocytes we could demonstrate that Dox induced Ca2+ dependent Ca2+ /calmodulin-dependent protein kinase II (CaMKII) activation is one of the factors involved in Dox cardiotoxicity by promoting apoptosis. Furthermore, we found that adeno-associated virus (AAV) mediated GRP78 overexpression partly protects neonatal cardiomyocytes from Dox induced cell death by modulating Ca2+ dependent pathways like the activation of CaMKII, phospholamban (PLN) and p53 accumulation. Most importantly, cardiac GRP78 gene therapy in mice treated with Dox revealed improved diastolic function (dP/dtmin) and survival after Dox treatment. In conclusion, our results demonstrate for the first time that Ca2+ dependent CaMKII activation fosters Dox cardiomyopathy and provide additional insight into possible mechanisms by which GRP78 overexpression protects cardiomyocytes from Doxorubicin toxicity.


Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cardiotoxicity/enzymology , Cardiotoxicity/pathology , Doxorubicin/toxicity , Heat-Shock Proteins/metabolism , Animals , Apoptosis/drug effects , Calcium/metabolism , Calcium Signaling/drug effects , Cardiotoxicity/physiopathology , Endoplasmic Reticulum Chaperone BiP , Enzyme Activation/drug effects , Homeostasis , Male , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , Phosphorylation , Rats , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/metabolism
7.
Toxicology ; 394: 72-83, 2018 02 01.
Article En | MEDLINE | ID: mdl-29248607

The tyrosine kinase inhibitor Sunitinib is used to treat cancer and is linked to severe adverse cardiovascular events. Mitogen activated kinase kinase 7 (MKK7) is involved in the development of cardiac injury and is a component of the c-Jun N-terminal kinase (JNK) signal transduction pathway. Apoptosis signal-regulating kinase 1 (ASK1) is the upstream activator of MKK7 and is specifically inhibited by 2,7-dihydro-2,7-dioxo-3H-naphtho[1,2,3-de]quinoline-1-carboxylic acid ethyl ester (NQDI-1). This study investigates the role of ASK1, MKK7 and JNK during Sunitinib-induced cardiotoxicity. Infarct size were measured in isolated male Sprague-Dawley rat Langendorff perfused hearts treated for 125 min with Sunitinib in the presence and absence of NQDI-1. Left ventricular cardiac tissue samples were analysed by qRT-PCR for MKK7 mRNA expression and cardiotoxicity associated microRNAs (miR-1, miR-27a, miR-133a and miR-133b) or Western blot analysis to measure ASK1/MKK7/JNK phosphorylation. Administration of Sunitinib (1 µM) during Langendorff perfusion resulted in increased infarct size, increased miR-133a expression, and decreased phosphorylation of the ASK1/MKK7/JNK pathway compared to control. Co-administration of NQDI-1 (2.5 µM) attenuated the increased Sunitinib-induced infarct size, reversed miR-133a expression and restored phosphorylated levels of ASK1/MKK7/JNK. These findings suggest that the ASK1/MKK7/JNK intracellular signalling pathway is important in Sunitinib-induced cardiotoxicity. The anti-cancer properties of Sunitinib were also assessed using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) cell viability assay. Sunitinib significantly decreased the cell viability of human acute myeloid leukemia 60 cell line (HL60). The combination of Sunitinib (1 nM-10 µM) with NQDI-1 (2.5 µM) enhanced the cancer-fighting properties of Sunitinib. Investigations into the ASK1/MKK7/JNK transduction pathway could lead to development of cardioprotective adjunct therapy, which could prevent Sunitinib-induced cardiac injury.


Cardiotoxicity/enzymology , Indoles/toxicity , MAP Kinase Kinase 7/metabolism , MAP Kinase Signaling System/drug effects , Pyrroles/toxicity , Animals , Aporphines/pharmacology , Cardiotoxicity/etiology , HL-60 Cells , Heart/drug effects , Humans , MAP Kinase Kinase 7/genetics , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/metabolism , Male , Myocardial Infarction/chemically induced , Myocardial Infarction/enzymology , Quinolines/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Sunitinib
8.
J Cell Mol Med ; 21(12): 3277-3287, 2017 Dec.
Article En | MEDLINE | ID: mdl-28608983

The use of doxorubicin (DOXO) as a chemotherapeutic drug has been hampered by cardiotoxicity leading to cardiomyopathy and heart failure. Folic acid (FA) is a modulator of endothelial nitric oxide (NO) synthase (eNOS), which in turn is an important player in diseases associated with NO insufficiency or NOS dysregulation, such as pressure overload and myocardial infarction. However, the role of FA in DOXO-induced cardiomyopathy is poorly understood. The aim of this study was to test the hypothesis that FA prevents DOXO-induced cardiomyopathy by modulating eNOS and mitochondrial structure and function. Male C57BL/6 mice were randomized to a single dose of DOXO (20 mg/kg intraperitoneal) or sham. FA supplementation (10 mg/day per oral) was started 7 days before DOXO injection and continued thereafter. DOXO resulted in 70% mortality after 10 days, with the surviving mice demonstrating a 30% reduction in stroke volume compared with sham groups. Pre-treatment with FA reduced mortality to 45% and improved stroke volume (both P < 0.05 versus DOXO). These effects of FA were underlain by blunting of DOXO-induced cardiomyocyte atrophy, apoptosis, interstitial fibrosis and impairment of mitochondrial function. Mechanistically, pre-treatment with FA prevented DOXO-induced increases in superoxide anion production by reducing the eNOS monomer:dimer ratio and eNOS S-glutathionylation, and attenuated DOXO-induced decreases in superoxide dismutase, eNOS phosphorylation and NO production. Enhancing eNOS function by restoring its coupling and subsequently reducing oxidative stress with FA may be a novel therapeutic approach to attenuate DOXO-induced cardiomyopathy.


Antioxidants/pharmacology , Cardiomyopathies/prevention & control , Cardiotonic Agents/pharmacology , Cardiotoxicity/prevention & control , Doxorubicin/antagonists & inhibitors , Doxorubicin/toxicity , Folic Acid/pharmacology , Animals , Antibiotics, Antineoplastic/toxicity , Apoptosis/drug effects , Cardiomyopathies/chemically induced , Cardiomyopathies/enzymology , Cardiomyopathies/mortality , Cardiotoxicity/enzymology , Cardiotoxicity/mortality , Cardiotoxicity/pathology , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/enzymology , Mitochondria/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Oxidative Stress/drug effects , Phosphorylation , Stroke Volume/drug effects , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxides/antagonists & inhibitors , Superoxides/metabolism , Survival Analysis
9.
Mol Med Rep ; 15(5): 2695-2702, 2017 May.
Article En | MEDLINE | ID: mdl-28447737

Doxorubicin (DOX) is one of the most effective chemotherapeutic agents for the treatment of a number of malignancies. However, its use is limited by serious cardiotoxic effects, for which there are currently no reliable pharmacologic therapies. Estrogen has exhibited protective effects against cardiac stressors in male and female animal models; however, its effects on DOX­induced cardiotoxicity remain unknown. High mortality and morbidity rates have been observed in patients with cancer worldwide, and DOX is often administered to a greater number of men than women. Therefore, the present study employed male Sprague-Dawley rats to evaluate the protective effects of 17ß-estradiol (E2) against DOX-induced cardiotoxicity. A total of 4 mg/kg DOX was administered to 14­week­old male Sprague­Dawley rats by intraperitoneal injection twice a week for 2 weeks. At 3 weeks following the first injection of DOX, an echocardiographic study revealed that DOX administration significantly decreased cardiac ejection fraction and fractional shortening by 20 and 29%, respectively, when compared with the vehicle­treated control rats (P<0.05). This was associated with decreased heart weight, myofibrillar disorganization and myofiber loss. The serum biomarkers for heart injury, including alanine aminotransferase, aspartate aminotransferase, lactate dehydrogenase and creatine kinase, were increased in DOX vs. vehicle­treated rats (P<0.05). E2 treatment by a daily subcutaneous injection of 2 mg/kg body weight attenuated the cardiotoxic effects of DOX. In addition, E2 treatment inhibited the DOX­induced increase in the expression of cardiac genes, nicotinamide adenine dinucleotide phosphate oxidase (NOX) 2, NOX4, B­cell lymphoma 2­associated X protein and caspase 3. These results demonstrate that E2 treatment may protect the heart against DOX-induced cardiotoxicity in male rats potentially through the regulation of NOX2, NOX4 and apoptosis genes.


Apoptosis/drug effects , Cardiotoxicity/prevention & control , Doxorubicin/adverse effects , Estradiol/pharmacology , NADPH Oxidase 2/metabolism , NADPH Oxidase 4/metabolism , Animals , Cardiotoxicity/enzymology , Cardiotoxicity/pathology , Doxorubicin/pharmacology , Female , Male , Rats , Rats, Sprague-Dawley
10.
Biol Sex Differ ; 8: 1, 2017.
Article En | MEDLINE | ID: mdl-28078076

BACKGROUND: There is inconclusive evidence about the role of sex as a risk factor for doxorubicin (DOX)-induced cardiotoxicity. Recent experimental studies have shown that adult female rats are protected against DOX-induced cardiotoxicity. However, the mechanisms of this sexual dimorphism are not fully elucidated. We have previously demonstrated that DOX alters the expression of several cytochrome P450 (CYP) enzymes in the hearts of male rats. Nevertheless, the sex-dependent effect of DOX on the expression of CYP enzymes is still not known. Therefore, in the present study, we determined the effect of acute DOX exposure on the expression of CYP genes in the hearts of both male and female C57Bl/6 mice. METHODS: Acute DOX cardiotoxicity was induced by a single intraperitoneal injection of 20 mg/kg DOX in male and female adult C57Bl/6 mice. Cardiac function was assessed 5 days after DOX exposure by trans-thoracic echocardiography. Mice were euthanized 1 day or 6 days after DOX or saline injection. Thereafter, the hearts were harvested and weighed. Heart sections were evaluated for pathological lesions. Total RNA was extracted and expression of natriuretic peptides, inflammatory and apoptotic markers, and CYP genes was measured by real-time PCR. RESULTS: Adult female C57Bl/6 mice were protected from acute DOX-induced cardiotoxicity as they show milder pathological lesions, less inflammation, and faster recovery from DOX-induced apoptosis and DOX-mediated inhibition of beta-type natriuretic peptide. Acute DOX exposure altered the gene expression of multiple CYP genes in a sex-dependent manner. In 24 h, DOX exposure caused male-specific induction of Cyp1b1 and female-specific induction of Cyp2c29 and Cyp2e1. CONCLUSIONS: Acute DOX exposure causes sex-dependent alteration of cardiac CYP gene expression. Since cardiac CYP enzymes metabolize several endogenous compounds to biologically active metabolites, sex-dependent alteration of CYP genes may play a role in the sexual dimorphism of acute DOX-induced cardiotoxicity.


Antibiotics, Antineoplastic/adverse effects , Cardiotoxicity/enzymology , Doxorubicin/adverse effects , Gene Expression Regulation, Enzymologic/drug effects , Myocardium/enzymology , Sex Characteristics , Animals , Atrial Natriuretic Factor/genetics , Cardiotoxicity/etiology , Cardiotoxicity/pathology , Cyclooxygenase 2/genetics , Cytochrome P-450 Enzyme System/genetics , Female , Heart/drug effects , Male , Mice, Inbred C57BL , Myocardium/pathology , Natriuretic Peptide, Brain/genetics , Proto-Oncogene Proteins c-bcl-2/genetics
11.
Arch Pharm Res ; 39(12): 1644-1652, 2016 Dec.
Article En | MEDLINE | ID: mdl-27681411

Astemizole, a non-sedating histamine H1 receptor blocker, is widely known to cause cardiac arrhythmia, which prolongs the QT interval. However, the precise molecular mechanism involved in antihistamine-induced cardiovascular adverse effects other than hERG channel inhibition is still unclear. In this study, we used DNA microarray analysis to detect the mechanisms involved in life-threatening adverse effects caused by astemizole. Rat primary cardiomyocytes were treated with various concentrations of astemizole for 24 h and the corresponding cell lysates were analyzed using a DNA microarray. Astemizole altered the expression profiles of genes involved in calcium transport/signaling. Using qRT-PCR analysis, we demonstrated that, among those genes, p21 (Cdc42/Rac)-activated kinase 1 (pak1) mRNA was downregulated by treatment with terfenadine and astemizole. Astemizole also reduced pak1 protein levels in rat cardiomyocytes. In addition, astemizole decreased pak1 mRNA and protein levels in H9c2 cells and induced an increase in cell surface area (hypertrophy) and cytotoxicity. Fingolimod hydrochloride (FTY720), a pak1 activator, inhibited astemizole-induced hypertrophy and cytotoxicity in H9c2 cells. These results suggest that antihistamine-induced cardiac adverse effects are associated with pak1 expression and function.


Histamine Antagonists/toxicity , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/enzymology , p21-Activated Kinases/metabolism , Animals , Cardiotoxicity/enzymology , Cells, Cultured , Dose-Response Relationship, Drug , Female , Histamine Antagonists/chemistry , Male , Pregnancy , Rats , Rats, Sprague-Dawley , p21-Activated Kinases/antagonists & inhibitors
12.
Br J Pharmacol ; 172(23): 5690-703, 2015 Dec.
Article En | MEDLINE | ID: mdl-25363561

BACKGROUND AND PURPOSE: The clinical use of doxorubicin, an effective anticancer drug, is severely hampered by its cardiotoxicity. 23-Hydroxybetulinic acid (23-HBA), isolated from Pulsatilla chinensis, enhances the anticancer effect of doxorubicin while simultaneously reducing its cardiac toxicity, but does not affect the concentration of doxorubicin in the plasma and heart. As the metabolite doxorubicinol is more potent than doxorubicin at inducing cardiac toxicity, in the present study we aimed to clarify the role of doxorubicinol in the protective effect of 23-HBA. EXPERIMENTAL APPROACH: Doxorubicin was administered to mice for two weeks in the presence or absence of 23-HBA. The heart pathology, function, myocardial enzymes and accumulation of doxorubicin and doxorubicinol were then analysed. A cellular pharmacokinetic study of doxorubicin and doxorubicinol, carbonyl reductase 1 (CBR1) interference and molecular docking was performed in vitro. KEY RESULTS: 23-HBA alleviated the doxorubicin-induced cardiotoxicity in mice, and this was accompanied by inhibition of the metabolism of doxorubicin and reduced accumulation of doxorubicinol selectively in hearts. In H9c2 cells, the protective effect of 23-HBA was shown to be closely associated with a decreased rate and extent of accumulation of doxorubicinol in mitochondria and nuclei. siRNA and docking analysis demonstrated that CBR1 has a crucial role in doxorubicin-mediated cardiotoxicity and 23-HBA inhibits this metabolic pathway. CONCLUSIONS AND IMPLICATIONS: Inhibition of CBR-mediated doxorubicin metabolism might be one of the protective mechanisms of 23-HBA against doxorubicin-induced cardiotoxicity. The present study provides a new research strategy guided by pharmacokinetic theory to elucidate the mechanism of drugs with unknown targets.


Alcohol Oxidoreductases/antagonists & inhibitors , Cardiotoxicity/prevention & control , Cytoprotection/drug effects , Doxorubicin/toxicity , Enzyme Inhibitors/pharmacology , Triterpenes/pharmacology , Alcohol Oxidoreductases/metabolism , Animals , Apoptosis/drug effects , Cardiotoxicity/enzymology , Cardiotoxicity/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Doxorubicin/adverse effects , Doxorubicin/analogs & derivatives , Enzyme Inhibitors/chemistry , Male , Mice , Mice, Inbred BALB C , Molecular Docking Simulation , Rats , Structure-Activity Relationship , Triterpenes/chemistry
13.
Cell Transplant ; 24(6): 1031-42, 2015.
Article En | MEDLINE | ID: mdl-24594448

Doxorubicin (DOX) is an effective chemotherapeutic drug used for the treatment of a variety of malignancies. Unfortunately, time and dose-dependent DOX therapy induces cardiotoxicity and heart failure. We previously reported that transplanted embryonic stem (ES) cells and the conditioned medium (CM) can repair and regenerate injured myocardium in acute DOX-induced cardiomyopathy (DIC). However, the effectiveness of ES cell and CM therapeutics has not been challenged in the chronic DIC model. To this end, the long-term impact of ES cells and CM on apoptosis, fibrosis, cytoplasmic vacuolization, oxidative stress, and their associated mediators were examined. Four weeks post-DIC, ES cells and CM-transplanted hearts showed a significant decrease in cardiac apoptotic nuclei, which was consequent to modulation of signaling molecules in the Akt pathway including PTEN, Akt, and mTOR. Cytoplasmic vacuolization was reduced following treatment with ES cells and CM, as was cardiac fibrosis, which was attributable to downregulation of MMP-9 activity. Oxidative stress, as evidenced by DHE staining and lipid peroxide concentration, was significantly diminished, and preservation of the antioxidant defense system was observed following CM and ES cell transplantation. In conclusion, our data suggest that transplanted ES cells and CM have long-term potentiation to significantly mitigate various adverse pathological mechanisms present in the injured chronic DIC heart.


Apoptosis , Cardiotoxicity/therapy , Doxorubicin/adverse effects , Mouse Embryonic Stem Cells/cytology , Proto-Oncogene Proteins c-akt/metabolism , Stem Cell Transplantation , TOR Serine-Threonine Kinases/metabolism , Animals , Antioxidants/metabolism , Apoptosis/drug effects , Cardiotoxicity/enzymology , Cardiotoxicity/pathology , Caspase 3/metabolism , Chronic Disease , Culture Media, Conditioned/pharmacology , Fibrosis , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Oxidative Stress/drug effects , Signal Transduction/drug effects , Vacuoles/drug effects , Vacuoles/metabolism
14.
Oxid Med Cell Longev ; 2014: 674219, 2014.
Article En | MEDLINE | ID: mdl-25386226

BACKGROUND: Doxorubicin, a potent chemotherapeutic agent, is associated with acute and chronic cardiotoxicity, which is cumulatively dose-dependent. Astragalus polysaccharide (APS), the extract of Astragalus membranaceus with strong antitumor and antiglomerulonephritis activity, can effectively alleviate inflammation. However, whether APS could ameliorate chemotherapy-induced cardiotoxicity is not understood. Here, we investigated the protective effects of APS on doxorubicin-induced cardiotoxicity and elucidated the underlying mechanisms of the protective effects of APS. METHODS: We analyzed myocardial injury in cancer patients who underwent doxorubicin chemotherapy and generated a doxorubicin-induced neonatal rat cardiomyocyte injury model and a mouse heart failure model. Echocardiography, reactive oxygen species (ROS) production, TUNEL, DNA laddering, and Western blotting were performed to observe cell survival, oxidative stress, and inflammatory signal pathways in cardiomyocytes. RESULTS: Treatment of patients with the chemotherapeutic drug doxorubicin led to heart dysfunction. Doxorubicin reduced cardiomyocyte viability and induced C57BL/6J mouse heart failure with concurrent elevated ROS generation and apoptosis, which, however, was attenuated by APS treatment. In addition, there was profound inhibition of p38MAPK and activation of Akt after APS treatment. CONCLUSIONS: These results demonstrate that APS could suppress oxidative stress and apoptosis, ameliorating doxorubicin-mediated cardiotoxicity by regulating the PI3k/Akt and p38MAPK pathways.


Astragalus Plant/metabolism , Cardiotoxicity/drug therapy , Cardiotoxicity/enzymology , Doxorubicin/toxicity , MAP Kinase Signaling System/drug effects , Polysaccharides/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cardiotoxicity/etiology , Doxorubicin/administration & dosage , Doxorubicin/adverse effects , Female , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Neoplasms/drug therapy , Phosphatidylinositol 3-Kinases/metabolism , Random Allocation , Rats , Rats, Sprague-Dawley , Retrospective Studies , p38 Mitogen-Activated Protein Kinases/metabolism
15.
Clin Cancer Res ; 20(18): 4873-81, 2014 Sep 15.
Article En | MEDLINE | ID: mdl-24714774

PURPOSE: Doxorubicin is one of the most effective chemotherapeutic agents. However, up to 30% of the patients treated with doxorubicin suffer from congestive heart failure. The mechanism of doxorubicin cardiotoxicity is likely multifactorial and most importantly, the genetic factors predisposing to doxorubicin cardiotoxicity are unknown. On the basis of the fact that mtDNA lesions and mitochondrial dysfunctions have been found in human hearts exposed to doxorubicin and that mitochondrial topoisomerase 1 (Top1mt) specifically controls mtDNA homeostasis, we hypothesized that Top1mt knockout (KO) mice might exhibit hypersensitivity to doxorubicin. EXPERIMENTAL DESIGN: Wild-type (WT) and KO Top1mt mice were treated once a week with 4 mg/kg doxorubicin for 8 weeks. Heart tissues were analyzed one week after the last treatment. RESULTS: Genetic inactivation of Top1mt in mice accentuates mtDNA copy number loss and mtDNA damage in heart tissue following doxorubicin treatment. Top1mt KO mice also fail to maintain respiratory chain protein production and mitochondrial cristae ultrastructure organization. These mitochondrial defects result in decreased O2 consumption, increased reactive oxygen species production, and enhanced heart muscle damage in animals treated with doxorubicin. Accordingly, Top1mt KO mice die within 45 days after the last doxorubicin injection, whereas the WT mice survive. CONCLUSIONS: Our results provide evidence that Top1mt, which is conserved across vertebrates, is critical for cardiac tolerance to doxorubicin and adaptive response to doxorubicin cardiotoxicity. They also suggest the potential of Top1mt single-nucleotide polymorphisms testing to investigate patient susceptibility to doxorubicin-induced cardiotoxicity.


Antibiotics, Antineoplastic/toxicity , Cardiotoxicity/enzymology , DNA Topoisomerases, Type I/metabolism , Doxorubicin/toxicity , Mitochondria, Heart/drug effects , Mitochondria, Heart/enzymology , Animals , Blotting, Western , DNA, Mitochondrial/drug effects , DNA, Mitochondrial/metabolism , Fluorescent Antibody Technique , Mice , Mice, Knockout
...