151
|
The Cardioprotective Role of N-Acetyl Cysteine Amide in the Prevention of Doxorubicin and Trastuzumab–Mediated Cardiac Dysfunction. Can J Cardiol 2016; 32:1513-1519. [DOI: 10.1016/j.cjca.2016.06.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 06/03/2016] [Accepted: 06/03/2016] [Indexed: 11/19/2022] Open
|
152
|
Elshazly SM, Mahmoud AA, Barakat W. Pentoxifylline abrogates cardiotoxicity induced by the administration of a single high dose or multiple low doses of doxorubicin in rats. Can J Physiol Pharmacol 2016; 94:1170-1177. [DOI: 10.1139/cjpp-2016-0115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Doxorubicin (DOX) possesses a broad-spectrum antineoplastic activity; however, its clinical application is impeded by cardiotoxicity. This study aimed to investigate the protective effect of pentoxifylline (PXF), which possesses antioxidant and anti-inflammatory properties against cardiotoxicity induced by a single high dose (15 mg/kg, i.p.) or multiple low doses (2.5 mg/kg, i.p., three times per week for 2 weeks) of DOX. At the end of the experimental period, the serum creatine kinase (CK)-MB and lactate dehydrogenase (LDH) activities were measured. The hearts were then removed for evaluating TNF-α, NO, malondialdehyde (MDA), and reduced glutathione (GSH) levels, superoxide dismutase (SOD) and catalase (CAT) activities, and the expression of iNOS, NF-κB, Fas ligand (FasL), and caspase-3. The administration of DOX in both dose regimens caused increases in serum CK-MB and LDH activities, in cardiac TNF-α, NO and MDA levels, as well as in the cardiac expression of iNOS, NF-κB, FasL and caspase-3, whereas it significantly reduced the cardiac GSH level, as well as SOD and CAT activities (P < 0.05). Prophylactic treatment of rats with PXF diminished DOX-induced alterations in theses parameters. Our results warrant the clinical use of PXF as an adjuvant therapy to abrogate cardiotoxicity of DOX and extend its clinical applications.
Collapse
Affiliation(s)
- Shimaa M. Elshazly
- Department of Pharmacology, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Amr A.A. Mahmoud
- Department of Pharmacology, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Waleed Barakat
- Department of Pharmacology, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
- Department of Pharmacology, Faculty of Pharmacy, Tabuk University, Tabuk 71491, Kingdom of Saudi Arabia
| |
Collapse
|
153
|
Carresi C, Gliozzi M, Giancotta C, Scarcella A, Scarano F, Bosco F, Mollace R, Tavernese A, Vitale C, Musolino V. Studies on the protective role of Bergamot polyphenols in doxorubicin-induced cardiotoxicity. PHARMANUTRITION 2016. [DOI: 10.1016/j.phanu.2015.11.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
154
|
Lee HJ, Kim DE, Park DJ, Choi GH, Yang DN, Heo JS, Lee SC. pH-Responsive mineralized nanoparticles as stable nanocarriers for intracellular nitric oxide delivery. Colloids Surf B Biointerfaces 2016; 146:1-8. [DOI: 10.1016/j.colsurfb.2016.05.039] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 05/12/2016] [Accepted: 05/14/2016] [Indexed: 01/05/2023]
|
155
|
Salimi A, Gholamifar E, Naserzadeh P, Hosseini MJ, Pourahmad J. Toxicity of lithium on isolated heart mitochondria and cardiomyocyte: A justification for its cardiotoxic adverse effect. J Biochem Mol Toxicol 2016; 31. [PMID: 27588890 DOI: 10.1002/jbt.21836] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Revised: 07/24/2016] [Accepted: 08/10/2016] [Indexed: 11/12/2022]
Abstract
Mitochondria play an important role in myocardial tissue homeostasis; therefore, deterioration in mitochondrial function will eventually lead to cardiomyocyte and endothelial cell death and consequently cardiovascular dysfunction. Lithium (Li+ ) is an effective drug for bipolar disorder with known cardiotoxic side effects. This study was designed to investigate the effects of Li+ on mitochondria and cardiomyocytes isolated from the heart of Wistar rat. Results revealed that Li+ induced a concentration- and time-dependent rise in mitochondrial ROS formation, inhibition of respiratory complexes (II), mitochondrial membrane potential (MMP) collapse, mitochondrial swelling, and cytochrome c release in rat heart mitochondria and also induced Caspase 3 activation through mitochondrial pathway, decline of ATP and lipid peroxidation in rat cardiomyocytes. These results indicate that the cardiotoxic effects of Li+ were initiated from mitochondrial dysfunction and oxidative stress, which finally ends in cytochrome c release and cell death signaling heart cardiomyocytes.
Collapse
Affiliation(s)
- Ahmad Salimi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Students Research Committee, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Pharmacology and Toxicology, School of Pharmacy, Ardabil University of Medical Science, Ardabil, Iran
| | - Ehsan Gholamifar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parvaneh Naserzadeh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Pharmacology and Toxicology, School of Pharmacy, Ardabil University of Medical Science, Ardabil, Iran
| | - Mir-Jamal Hosseini
- Zanjan Applied Pharmacology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.,Department of Pharmacology and Toxicology, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Jalal Pourahmad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
156
|
Wu J, Guo W, Lin SZ, Wang ZJ, Kan JT, Chen SY, Zhu YZ. Gp130-mediated STAT3 activation by S-propargyl-cysteine, an endogenous hydrogen sulfide initiator, prevents doxorubicin-induced cardiotoxicity. Cell Death Dis 2016; 7:e2339. [PMID: 27537522 PMCID: PMC5108313 DOI: 10.1038/cddis.2016.209] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 06/08/2016] [Accepted: 06/09/2016] [Indexed: 12/20/2022]
Abstract
Doxorubicin (Dox) could trigger a large amount of apoptotic cells in the myocardium, which leads to dilated cardiomyopathy and heart failure. S-propargyl-cysteine (SPRC), a producing agent of endogenous hydrogen sulfide (H2S), possesses cardioprotective efficacy. However, the specific effect and mechanism of SPRC in Dox-induced cardiotoxicity remain elusive. Given gp130 with its main downstream signaling molecule, signal transducer and activator of transcription 3 (STAT3), is involved in cardiac myocyte survival and growth; the present study was performed to elucidate whether SPRC counteracts Dox-induced cardiotoxicity, and if so, whether the gp130/STAT3 pathway is involved in this cardioprotective activity. SPRC stimulated the activation of STAT3 via gp130-mediated transduction tunnel in vitro and in vivo. In Dox-stimulated cardiotoxicity, SPRC enhanced cell viability, restored expression of gp130/STAT3-regulated downstream genes, inhibited apoptosis and oxidative stress, and antagonized mitochondrial dysfunction and intracellular Ca(2+) overload. Intriguingly, blockade of gp130/STAT3 signaling abrogated all these beneficial capacities of SPRC. Our findings present the first piece of evidence for the therapeutic properties of SPRC in alleviating Dox cardiotoxicity, which could be attributed to the activation of gp130-mediated STAT3 signaling. This will offer a novel molecular basis and therapeutic strategy of H2S donor for the treatment of heart failure.
Collapse
Affiliation(s)
- J Wu
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - W Guo
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - S-Z Lin
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Z-J Wang
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - J-T Kan
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - S-Y Chen
- Department of Cardiovascular Surgery, Guangdong General Hospital, Guangzhou, Guangdong, China
| | - Y-Z Zhu
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China.,School of Pharmacy, Macau University of Science & Technology, Macau, China
| |
Collapse
|
157
|
Ezzat SM, El Gaafary M, El Sayed AM, Sabry OM, Ali ZY, Hafner S, Schmiech M, Jin L, Syrovets T, Simmet T. The Cardenolide Glycoside Acovenoside A Affords Protective Activity in Doxorubicin-Induced Cardiotoxicity in Mice. J Pharmacol Exp Ther 2016; 358:262-70. [PMID: 27247000 DOI: 10.1124/jpet.116.232652] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Accepted: 11/15/2011] [Indexed: 03/08/2025] Open
Abstract
The current study aimed to investigate the protective effect of the cardenolide glycoside acovenoside A (AcoA) against doxorubicin-induced cardiotoxicity in mice. AcoA was isolated from the pericarps of Acokanthera oppositifolia to chemical homogeneity and characterized by means of one- and two-dimensional nuclear magnetic resonance spectroscopy. AcoA exhibited relatively low toxicity in mice (LD50 = 223.3 mg/kg bw). Repeated administration of doxorubicin induced cardiotoxicity manifested by reduced activity of myocardial membrane-bound ion pumps and elevated serum biomarkers of myocardial dysfunction, oxidative stress, and inflammation. Pretreatment of doxorubicin-exposed mice with AcoA (11.16 or 22.33 mg/kg bw, i.p.) for 2 weeks after 2 weeks of combined administration of AcoA and doxorubicin protected the animals dose dependently against doxorubicin-induced cardiotoxicity as indicated by normalization of the levels of different myocardial markers of oxidative stress (malondialdehyde, nitric oxide, total antioxidant capacity, and cardiac glutathione), serum myocardial diagnostic marker enzymes (serum cardiac troponin T, creatine kinase isoenzyme MB, aspartate aminotransferase, and lactate dehydrogenase), and inflammatory markers (c-reactive protein, tumor necrosis factor-α, and interleukin-6), as well as myocardial Na(+)/K(+)-ATPase activity. These effects were attributed to the negative impact of AcoA on transcription factors nuclear factor κB and interferon regulatory factor 3/7. Thus acovenoside A might act as a cardioprotective agent to prevent doxorubicin-induced cardiotoxicity.
Collapse
Affiliation(s)
- Shahira M Ezzat
- Department of Pharmacognosy, College of Pharmacy, Cairo University, Giza, Egypt (S.M.E., M.E.G., A.M.E.S., O.M.S.); Department of Biochemistry, National Organization for Drug Control and Research (NODCAR), Cairo, Egypt (Z.Y.A.); Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, D-89081 Ulm, Germany (M.E.G., S.H., M.S., L.J., Ta.S., Th.S.)
| | - Menna El Gaafary
- Department of Pharmacognosy, College of Pharmacy, Cairo University, Giza, Egypt (S.M.E., M.E.G., A.M.E.S., O.M.S.); Department of Biochemistry, National Organization for Drug Control and Research (NODCAR), Cairo, Egypt (Z.Y.A.); Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, D-89081 Ulm, Germany (M.E.G., S.H., M.S., L.J., Ta.S., Th.S.)
| | - Abeer M El Sayed
- Department of Pharmacognosy, College of Pharmacy, Cairo University, Giza, Egypt (S.M.E., M.E.G., A.M.E.S., O.M.S.); Department of Biochemistry, National Organization for Drug Control and Research (NODCAR), Cairo, Egypt (Z.Y.A.); Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, D-89081 Ulm, Germany (M.E.G., S.H., M.S., L.J., Ta.S., Th.S.)
| | - Omar M Sabry
- Department of Pharmacognosy, College of Pharmacy, Cairo University, Giza, Egypt (S.M.E., M.E.G., A.M.E.S., O.M.S.); Department of Biochemistry, National Organization for Drug Control and Research (NODCAR), Cairo, Egypt (Z.Y.A.); Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, D-89081 Ulm, Germany (M.E.G., S.H., M.S., L.J., Ta.S., Th.S.)
| | - Zeinab Y Ali
- Department of Pharmacognosy, College of Pharmacy, Cairo University, Giza, Egypt (S.M.E., M.E.G., A.M.E.S., O.M.S.); Department of Biochemistry, National Organization for Drug Control and Research (NODCAR), Cairo, Egypt (Z.Y.A.); Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, D-89081 Ulm, Germany (M.E.G., S.H., M.S., L.J., Ta.S., Th.S.)
| | - Susanne Hafner
- Department of Pharmacognosy, College of Pharmacy, Cairo University, Giza, Egypt (S.M.E., M.E.G., A.M.E.S., O.M.S.); Department of Biochemistry, National Organization for Drug Control and Research (NODCAR), Cairo, Egypt (Z.Y.A.); Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, D-89081 Ulm, Germany (M.E.G., S.H., M.S., L.J., Ta.S., Th.S.)
| | - Michael Schmiech
- Department of Pharmacognosy, College of Pharmacy, Cairo University, Giza, Egypt (S.M.E., M.E.G., A.M.E.S., O.M.S.); Department of Biochemistry, National Organization for Drug Control and Research (NODCAR), Cairo, Egypt (Z.Y.A.); Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, D-89081 Ulm, Germany (M.E.G., S.H., M.S., L.J., Ta.S., Th.S.)
| | - Lu Jin
- Department of Pharmacognosy, College of Pharmacy, Cairo University, Giza, Egypt (S.M.E., M.E.G., A.M.E.S., O.M.S.); Department of Biochemistry, National Organization for Drug Control and Research (NODCAR), Cairo, Egypt (Z.Y.A.); Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, D-89081 Ulm, Germany (M.E.G., S.H., M.S., L.J., Ta.S., Th.S.)
| | - Tatiana Syrovets
- Department of Pharmacognosy, College of Pharmacy, Cairo University, Giza, Egypt (S.M.E., M.E.G., A.M.E.S., O.M.S.); Department of Biochemistry, National Organization for Drug Control and Research (NODCAR), Cairo, Egypt (Z.Y.A.); Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, D-89081 Ulm, Germany (M.E.G., S.H., M.S., L.J., Ta.S., Th.S.)
| | - Thomas Simmet
- Department of Pharmacognosy, College of Pharmacy, Cairo University, Giza, Egypt (S.M.E., M.E.G., A.M.E.S., O.M.S.); Department of Biochemistry, National Organization for Drug Control and Research (NODCAR), Cairo, Egypt (Z.Y.A.); Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, D-89081 Ulm, Germany (M.E.G., S.H., M.S., L.J., Ta.S., Th.S.)
| |
Collapse
|
158
|
Bartolomeu AR, Frión-Herrera Y, da Silva LM, Romagnoli GG, de Oliveira DE, Sforcin JM. Combinatorial effects of geopropolis produced by Melipona fasciculata Smith with anticancer drugs against human laryngeal epidermoid carcinoma (HEp-2) cells. Biomed Pharmacother 2016; 81:48-55. [DOI: 10.1016/j.biopha.2016.03.049] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 03/30/2016] [Accepted: 03/31/2016] [Indexed: 01/13/2023] Open
|
159
|
The Role of Oxidative Stress in Myocardial Ischemia and Reperfusion Injury and Remodeling: Revisited. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:1656450. [PMID: 27313825 PMCID: PMC4897712 DOI: 10.1155/2016/1656450] [Citation(s) in RCA: 220] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 04/11/2016] [Accepted: 05/03/2016] [Indexed: 01/11/2023]
Abstract
Oxidative and reductive stress are dual dynamic phases experienced by the cells undergoing adaptation towards endogenous or exogenous noxious stimulus. The former arises due to the imbalance between the reactive oxygen species production and antioxidant defenses, while the latter is due to the aberrant increase in the reducing equivalents. Mitochondrial malfunction is the common denominator arising from the aberrant functioning of the rheostat that maintains the homeostasis between oxidative and reductive stress. Recent experimental evidences suggest that the maladaptation during oxidative stress could play a pivotal role in the pathophysiology of major cardiovascular diseases such as myocardial infraction, atherosclerosis, and diabetic cardiovascular complications. In this review we have discussed the role of oxidative and reductive stress pathways in the pathogenesis of myocardial ischemia/reperfusion injury and diabetic cardiomyopathy (DCM). Furthermore, we have provided impetus for the development of subcellular organelle targeted antioxidant drug therapy for thwarting the deterioration of the failing myocardium in the aforementioned cardiovascular conditions.
Collapse
|
160
|
Dębowska K, Dębski D, Michałowski B, Dybala-Defratyka A, Wójcik T, Michalski R, Jakubowska M, Selmi A, Smulik R, Piotrowski Ł, Adamus J, Marcinek A, Chlopicki S, Sikora A. Characterization of Fluorescein-Based Monoboronate Probe and Its Application to the Detection of Peroxynitrite in Endothelial Cells Treated with Doxorubicin. Chem Res Toxicol 2016; 29:735-46. [PMID: 27081868 DOI: 10.1021/acs.chemrestox.5b00431] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Boronate probes have emerged recently as a versatile tool for the detection of reactive oxygen and nitrogen species. Here, we present the characterization of a fluorescein-based monoboronate probe, a 4-(pinacol boronate)benzyl derivative of fluorescein methyl ester (FBBE), that proved to be useful to detect peroxynitrite in cell culture experiments. The reactivity of FBBE toward peroxynitrite as well hypochlorite, hydrogen peroxide, and tyrosyl hydroperoxide was determined. Second-order rate constants of the reactions of FBBE with peroxynitrite, HOCl, and H2O2 at pH 7.4 were equal to (2.8 ± 0.2) × 10(5) M(-1) s(-1), (8.6 ± 0.5) × 10(3) M(-1) s(-1), and (0.96 ± 0.03) M(-1) s(-1), respectively. The presence of glutathione completely blocked the oxidation of the probe by HOCl and significantly inhibited its oxidation by H2O2 and tyrosyl hydroperoxide but not by peroxynitrite. The oxidative conversion of the probe was also studied in the systems generating singlet oxygen, superoxide radical anion, and nitric oxide in the presence and absence of glutathione. Spectroscopic characterization of FBBE and its oxidation product has been also performed. The differences in the reactivity pattern were supported by DFT quantum mechanical calculations. Finally, the FBBE probe was used to study the oxidative stress in endothelial cells (Ea.hy926) incubated with doxorubicin, a quinone anthracycline antibiotic. In endothelial cells pretreated with doxorubicin, FBBE was oxidized, and this effect was reversed by PEG-SOD and L-NAME but not by catalase.
Collapse
Affiliation(s)
- Karolina Dębowska
- Institute of Applied Radiation Chemistry, Lodz University of Technology , Lodz, Poland
| | - Dawid Dębski
- Institute of Applied Radiation Chemistry, Lodz University of Technology , Lodz, Poland
| | - Bartosz Michałowski
- Institute of Applied Radiation Chemistry, Lodz University of Technology , Lodz, Poland
| | | | - Tomasz Wójcik
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University , Kraków, Poland
| | - Radosław Michalski
- Institute of Applied Radiation Chemistry, Lodz University of Technology , Lodz, Poland
| | - Małgorzata Jakubowska
- Institute of Applied Radiation Chemistry, Lodz University of Technology , Lodz, Poland
| | - Anna Selmi
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University , Kraków, Poland
| | - Renata Smulik
- Institute of Applied Radiation Chemistry, Lodz University of Technology , Lodz, Poland
| | - Łukasz Piotrowski
- Institute of Applied Radiation Chemistry, Lodz University of Technology , Lodz, Poland
| | - Jan Adamus
- Institute of Applied Radiation Chemistry, Lodz University of Technology , Lodz, Poland
| | - Andrzej Marcinek
- Institute of Applied Radiation Chemistry, Lodz University of Technology , Lodz, Poland
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University , Kraków, Poland.,Chair of Pharmacology, Jagiellonian University Medical College , Kraków, Poland
| | - Adam Sikora
- Institute of Applied Radiation Chemistry, Lodz University of Technology , Lodz, Poland
| |
Collapse
|
161
|
Role of TFEB Mediated Autophagy, Oxidative Stress, Inflammation, and Cell Death in Endotoxin Induced Myocardial Toxicity of Young and Aged Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:5380319. [PMID: 27200146 PMCID: PMC4856916 DOI: 10.1155/2016/5380319] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 03/25/2016] [Accepted: 04/03/2016] [Indexed: 11/25/2022]
Abstract
Elderly patients are susceptible to sepsis. LPS induced myocardial injury is a widely used animal model to assess sepsis induced cardiac dysfunction. The age dependent mechanisms behind sepsis susceptibility were not studied. We analyzed age associated changes to cardiac function, cell death, inflammation, oxidative stress, and autophagy in LPS induced myocardial injury. Both young and aged C57BL/6 mice were used for LPS administration. The results demonstrated that LPS induced more cardiac injury (creatine kinase, lactate dehydrogenase, troponin I, and cardiac myosin-light chains 1), cardiac dysfunction (left ventricular inner dimension, LVID, and ejection fraction (EF)), cell death, inflammation, and oxidative stress in aged mice compared to young mice. However, a significant age dependent decline in autophagy was observed. Translocation of Transcription Factor EB (TFEB) to nucleus and formation of LC3-II were significantly reduced in LPS administered aged mice compared to young ones. In addition to that, downstream effector of TFEB, LAMP-1, was induced in response to LPS challenge in young mice. The present study newly demonstrates that TFEB mediated autophagy is crucial for protection against LPS induced myocardial injury particularly in aging senescent heart. Targeting this autophagy-oxidative stress-inflammation-cell death axis may provide a novel therapeutic strategy for cardioprotection in the elderly.
Collapse
|
162
|
Liguzinediol protects against cardiac fibrosis in rats in vivo and in vitro. Biomed Pharmacother 2016; 80:260-267. [PMID: 27133065 DOI: 10.1016/j.biopha.2016.03.033] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 03/25/2016] [Indexed: 11/22/2022] Open
Abstract
Cardiac fibrosis plays a causal role in the development of heart failure, and anti-fibrotic therapy represents a promising strategy to mitigate heart failure. The purpose of this study was to investigate the effect of a new drug-liguzinediol on cardiac fibrosis of heart failure Male Sprague-Dawley rats (SD) rats and the underlying mechanisms. Liguzinediol was administered to rats that were injected with doxorubicin (Dox) for four weeks. Two weeks later, its effects on cardiac fibrosis were assessed by haematoxylin and eosin (HE) staining and Masson staining. The collagen content was determined by Elisa, and protein expression was detected by western blot in vitro and in vivo. Liguzinediol decreased cardiac muscle fiber break evidenced by HE staining and it significantly reduced cardiac fibrosis evidenced by Masson staining in DOX-treated rats. In addition, the hydroxyproline level and the ratio of type I/III collagens were also significantly decreased, and western blot assays showed that liguzinediol regulated the balance between matrix matalloproteinases (MMPs) and tissue inhibitor of metalloproteinase (TIMPs) to protect cardiac remodeling in vivo and in vitro. These data collectively indicated that liguzinediol could protect against cardiac fibrosis in rats. Liguzinediol could be exploited to be a promising candidate for cardiac fibrosis.
Collapse
|
163
|
Somasundaram V, Nadhan R, K Hemalatha S, Kumar Sengodan S, Srinivas P. Nitric oxide and reactive oxygen species: Clues to target oxidative damage repair defective breast cancers. Crit Rev Oncol Hematol 2016; 101:184-92. [PMID: 27017408 DOI: 10.1016/j.critrevonc.2016.03.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 01/19/2016] [Accepted: 03/02/2016] [Indexed: 12/21/2022] Open
Abstract
The identification of various biomolecules in cancer progression and therapy has led to the exploration of the roles of two cardinal players, namely Nitric Oxide (NO) and Reactive Oxygen Species (ROS) in cancer. Both ROS and NO display bimodal fashions of functional activity in a concentration dependent manner, by inducing either pro- or anti- tumorigenic signals. Researchers have identified the potential capability of NO and ROS in therapies owing to their role in eliciting pro-apoptotic signals at higher concentrations and their ability to sensitize cancer cells to one another as well as to other therapeutics. We review the prospects of NO and ROS in cancer progression and therapy, and analyze the role of a combinatorial therapy wherein an NO donor (SNAP) is used to sensitize the oxidative damage repair defective, triple negative breast cancer cells (HCC 1937) to a potent ROS inducer. Preliminary findings support the potential to employ various combinatorial regimes for anti-cancer therapies with regard to exploiting the chemo-sensitization property of NO donors.
Collapse
Affiliation(s)
- Veena Somasundaram
- Cancer Research Program 5, Rajiv Gandhi Centre for Biotechnology, Thycaud P O, Poojappura, Thiruvananthapuram 695 014, Kerala, India
| | - Revathy Nadhan
- Cancer Research Program 5, Rajiv Gandhi Centre for Biotechnology, Thycaud P O, Poojappura, Thiruvananthapuram 695 014, Kerala, India
| | - Sreelatha K Hemalatha
- Cancer Research Program 5, Rajiv Gandhi Centre for Biotechnology, Thycaud P O, Poojappura, Thiruvananthapuram 695 014, Kerala, India
| | - Satheesh Kumar Sengodan
- Cancer Research Program 5, Rajiv Gandhi Centre for Biotechnology, Thycaud P O, Poojappura, Thiruvananthapuram 695 014, Kerala, India
| | - Priya Srinivas
- Cancer Research Program 5, Rajiv Gandhi Centre for Biotechnology, Thycaud P O, Poojappura, Thiruvananthapuram 695 014, Kerala, India.
| |
Collapse
|
164
|
Zielonka J, Zielonka M, VerPlank L, Cheng G, Hardy M, Ouari O, Ayhan MM, Podsiadły R, Sikora A, Lambeth JD, Kalyanaraman B. Mitigation of NADPH Oxidase 2 Activity as a Strategy to Inhibit Peroxynitrite Formation. J Biol Chem 2016; 291:7029-44. [PMID: 26839313 DOI: 10.1074/jbc.m115.702787] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Indexed: 01/09/2023] Open
Abstract
Using high throughput screening-compatible assays for superoxide and hydrogen peroxide, we identified potential inhibitors of the NADPH oxidase (Nox2) isoform from a small library of bioactive compounds. By using multiple probes (hydroethidine, hydropropidine, Amplex Red, and coumarin boronate) with well defined redox chemistry that form highly diagnostic marker products upon reaction with superoxide (O2 (̇̄)), hydrogen peroxide (H2O2), and peroxynitrite (ONOO(-)), the number of false positives was greatly decreased. Selected hits for Nox2 were further screened for their ability to inhibit ONOO(-)formation in activated macrophages. A new diagnostic marker product for ONOO(-)is reported. We conclude that the newly developed high throughput screening/reactive oxygen species assays could also be used to identify potential inhibitors of ONOO(-)formed from Nox2-derived O2 (̇̄)and nitric oxide synthase-derived nitric oxide.
Collapse
Affiliation(s)
- Jacek Zielonka
- From the Department of Biophysics and Free Radical Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin 53226,
| | - Monika Zielonka
- From the Department of Biophysics and Free Radical Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Lynn VerPlank
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142
| | - Gang Cheng
- From the Department of Biophysics and Free Radical Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Micael Hardy
- the Aix-Marseille Université, CNRS, ICR UMR 7273, 13397 Marseille, France
| | - Olivier Ouari
- the Aix-Marseille Université, CNRS, ICR UMR 7273, 13397 Marseille, France
| | - Mehmet Menaf Ayhan
- the Aix-Marseille Université, CNRS, ICR UMR 7273, 13397 Marseille, France
| | - Radosław Podsiadły
- From the Department of Biophysics and Free Radical Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Adam Sikora
- the Institute of Applied Radiation Chemistry, Faculty of Chemistry, Lodz University of Technology, Żeromskiego 116, 90-924 Lodz, Poland, and
| | - J David Lambeth
- the Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia 30322
| | - Balaraman Kalyanaraman
- From the Department of Biophysics and Free Radical Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin 53226,
| |
Collapse
|
165
|
New signal transduction paradigms in anthracycline-induced cardiotoxicity. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:1916-25. [PMID: 26828775 DOI: 10.1016/j.bbamcr.2016.01.021] [Citation(s) in RCA: 146] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 01/06/2016] [Accepted: 01/28/2016] [Indexed: 12/21/2022]
Abstract
Anthracyclines, such as doxorubicin, are the most potent and widely used chemotherapeutic agents for the treatment of a variety of human cancers, including solid tumors and hematological malignancies. However, their clinical use is hampered by severe cardiotoxic side effects and cancer therapy-related heart disease has become a leading cause of morbidity and mortality among cancer survivors. The identification of therapeutic strategies limiting anthracycline cardiotoxicity with preserved antitumor efficacy thus represents the current challenge of cardio-oncologists. Anthracycline cardiotoxicity has been originally ascribed to the ability of this class of drugs to disrupt iron metabolism and generate excess of reactive oxygen species (ROS). However, small clinical trials with iron chelators and anti-oxidants failed to provide any benefit and suggested that doxorubicin cardiotoxicity is not solely due to redox cycling. New emerging explanations include anthracycline-dependent regulation of major signaling pathways controlling DNA damage response, cardiomyocyte survival, cardiac inflammation, energetic stress and gene expression modulation. This review will summarize recent studies unraveling the complex web of mechanisms of doxorubicin-mediated cardiotoxicity, and identifying new druggable players for the prevention of heart disease in cancer patients. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Integration of Developmental and Environmental Cues in the Heart edited by Marcus Schaub and Hughes Abriel.
Collapse
|
166
|
Zhang C, Wu Y, Dong Y, Xu H, Zhao L. Quantification of DOX bioavailability in biological samples of mice by sensitive and precise HPLC assay. PHARMACEUTICAL BIOLOGY 2016; 54:55-61. [PMID: 25880143 DOI: 10.3109/13880209.2015.1014918] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
CONTEXT Doxorubicin (DOX)-loaded folate-targeted poly(3-hydroxybutyrate-co-3-hydroxyoctanoate) [P(HB-HO)] nanoparticles [DOX/FA-PEG-P(HB-HO) NPs] were prepared by the W1/O/W2 solvent extraction/evaporation method for applications in cancer treatment. However, the biodistribution, pharmacokinetics, and targeting of the nanoparticles (NPs) have not yet been studied. OBJECTIVE The biodistribution, pharmacokinetics, and targeting of DOX/FA-PEG-P(HB-HO) NPs were evaluated in female BALB/c nude mice bearing HeLa tumors. MATERIALS AND METHODS Three DOX formulations were injected into the tail vein of the mice at a dosage of 5 mg/kg. At each time point, blood and various tissues were collected. All samples were then processed and analyzed by a validated high performance liquid chromatographic (HPLC) method. RESULTS The t1/2 values of DOX/P(HB-HO) NPs and DOX/FA-PEG-P(HB-HO) NPs were 2.7- and 3.5-times higher than that of free DOX. No significant difference (p > 0.05) was found in Cmax between the NPs and free DOX. The Tmax values of the two NPs were prolonged from 0.25 to 1 h. The AUC0-t values were 1.55- and 3.05-folds higher than that of free DOX, and MRT increased to 15.99 h for DOX/P(HB-HO) NPs and 25.14 h for DOX/FA-PEG-P(HB-HO) NPs. For DOX/FA-PEG-P(HB-HO) NPs, the DOX content in the tumors were 10.81- and 3.33-times higher than those for free DOX and DOX/P(HB-HO) NPs at 48 h, respectively. DISCUSSION AND CONCLUSIONS DOX/FA-PEG-P(HB-HO) NPs displayed reduced cardiac toxicity and improved bioavailability. Moreover, the NPs exhibited a significant extent of DOX accumulation in the tumors, thus suggesting that folate-targeted NPs could effectively transport into HeLa tumors with satisfying targeting.
Collapse
MESH Headings
- Animals
- Antibiotics, Antineoplastic/administration & dosage
- Antibiotics, Antineoplastic/blood
- Antibiotics, Antineoplastic/chemistry
- Antibiotics, Antineoplastic/pharmacokinetics
- Antibiotics, Antineoplastic/toxicity
- Area Under Curve
- Biological Availability
- Cardiotoxicity
- Chemistry, Pharmaceutical
- Chromatography, High Pressure Liquid
- Doxorubicin/administration & dosage
- Doxorubicin/blood
- Doxorubicin/chemistry
- Doxorubicin/pharmacokinetics
- Doxorubicin/toxicity
- Drug Carriers
- Drug Monitoring/methods
- Female
- Folic Acid/chemistry
- Folic Acid/metabolism
- Half-Life
- HeLa Cells
- Heart Diseases/chemically induced
- Humans
- Injections, Intravenous
- Metabolic Clearance Rate
- Mice, Inbred BALB C
- Mice, Nude
- Nanoparticles
- Polyesters/chemistry
- Risk Assessment
- Tissue Distribution
- Uterine Cervical Neoplasms/drug therapy
- Uterine Cervical Neoplasms/metabolism
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Chan Zhang
- a School of Environment and Safety, Taiyuan University of Science and Technology , Taiyuan , China
| | - Yifan Wu
- b College of Life Science, Shanxi University , Taiyuan , China , and
| | - Yuefeng Dong
- c Shanxi Institute of Medicine and Life Science , Taiyuan , China
| | - Hongying Xu
- a School of Environment and Safety, Taiyuan University of Science and Technology , Taiyuan , China
| | - Liangqi Zhao
- b College of Life Science, Shanxi University , Taiyuan , China , and
| |
Collapse
|
167
|
Li L, Li Q, Chen P, Li Z, Chen Z, Tang B. Consecutive Gated Injection-Based Microchip Electrophoresis for Simultaneous Quantitation of Superoxide Anion and Nitric Oxide in Single PC-12 Cells. Anal Chem 2015; 88:930-6. [DOI: 10.1021/acs.analchem.5b03664] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Lu Li
- College of Chemistry,
Chemical
Engineering and Materials Science, Collaborative Innovation Center
of Functionalized Probes for Chemical Imaging in Universities of Shandong,
Key Laboratory of Molecular and Nano Probes, Ministry
of Education, Shandong Normal University, Jinan, 250014, P.R. China
| | - Qingling Li
- College of Chemistry,
Chemical
Engineering and Materials Science, Collaborative Innovation Center
of Functionalized Probes for Chemical Imaging in Universities of Shandong,
Key Laboratory of Molecular and Nano Probes, Ministry
of Education, Shandong Normal University, Jinan, 250014, P.R. China
| | - Peilin Chen
- College of Chemistry,
Chemical
Engineering and Materials Science, Collaborative Innovation Center
of Functionalized Probes for Chemical Imaging in Universities of Shandong,
Key Laboratory of Molecular and Nano Probes, Ministry
of Education, Shandong Normal University, Jinan, 250014, P.R. China
| | - Zhongyi Li
- College of Chemistry,
Chemical
Engineering and Materials Science, Collaborative Innovation Center
of Functionalized Probes for Chemical Imaging in Universities of Shandong,
Key Laboratory of Molecular and Nano Probes, Ministry
of Education, Shandong Normal University, Jinan, 250014, P.R. China
| | - Zhenzhen Chen
- College of Chemistry,
Chemical
Engineering and Materials Science, Collaborative Innovation Center
of Functionalized Probes for Chemical Imaging in Universities of Shandong,
Key Laboratory of Molecular and Nano Probes, Ministry
of Education, Shandong Normal University, Jinan, 250014, P.R. China
| | - Bo Tang
- College of Chemistry,
Chemical
Engineering and Materials Science, Collaborative Innovation Center
of Functionalized Probes for Chemical Imaging in Universities of Shandong,
Key Laboratory of Molecular and Nano Probes, Ministry
of Education, Shandong Normal University, Jinan, 250014, P.R. China
| |
Collapse
|
168
|
Cappetta D, Esposito G, Piegari E, Russo R, Ciuffreda LP, Rivellino A, Berrino L, Rossi F, De Angelis A, Urbanek K. SIRT1 activation attenuates diastolic dysfunction by reducing cardiac fibrosis in a model of anthracycline cardiomyopathy. Int J Cardiol 2015; 205:99-110. [PMID: 26730840 DOI: 10.1016/j.ijcard.2015.12.008] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 12/04/2015] [Accepted: 12/12/2015] [Indexed: 02/08/2023]
Abstract
BACKGROUND Doxorubicin (DOXO) is an effective anti-neoplastic drug but its clinical benefits are hampered by cardiotoxicity. Oxidative stress, apoptosis and myocardial fibrosis mediate the anthracycline cardiomyopathy. ROS trigger TGF-β pathway that activates cardiac fibroblasts promoting fibrosis. Myocardial stiffness contributes to diastolic dysfunction, less studied aspect of anthracycline cardiomyopathy. Considering the role of SIRT1 in the inhibition of the TGF-β/SMAD3 pathway, resveratrol (RES), a SIRT1 activator, might improve cardiac function by interfering with the development of cardiac fibrosis in a model of DOXO-induced cardiomyopathy. METHODS F344 rats received a cumulative dose of 15 mg/kg of DOXO in 2 weeks or DOXO+RES (DOXO and RES, 2.5mg/kg/day, concomitantly for 2 weeks and then RES alone for 1 more week). The effects of RES on cardiac fibroblasts were also tested in vitro. RESULTS Along with systolic dysfunction, DOXO was also responsible of diastolic abnormalities. Myocardial stiffness correlated with fibroblast activation and collagen deposition. DOXO+RES co-treatment significantly improved ± dP/dt and, more interestingly, ameliorated end-diastolic pressure/volume relationship. Treatment with RES resulted in reduced fibrosis and fibroblast activation and, most importantly, the mortality rate was significantly reduced in DOXO+RES group. Fibroblasts isolated from DOXO+RES-treated rats, in which SIRT1 was upregulated, showed decreased levels of TGF-β and pSMAD3/SMAD3 when compared to cells isolated from DOXO-exposed hearts. CONCLUSIONS Our findings reveal a key role of SIRT1 in supporting animal survival and functional parameters of the heart. SIRT1 activation by interfering with fibrogenesis can improve relaxation properties of myocardium and attenuate myocardial remodeling related to chemotherapy.
Collapse
Affiliation(s)
- Donato Cappetta
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Via Costantinopoli 16, 80138 Naples, Italy
| | - Grazia Esposito
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Via Costantinopoli 16, 80138 Naples, Italy
| | - Elena Piegari
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Via Costantinopoli 16, 80138 Naples, Italy
| | - Rosa Russo
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Via Costantinopoli 16, 80138 Naples, Italy
| | - Loreta Pia Ciuffreda
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Via Costantinopoli 16, 80138 Naples, Italy
| | - Alessia Rivellino
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Via Costantinopoli 16, 80138 Naples, Italy
| | - Liberato Berrino
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Via Costantinopoli 16, 80138 Naples, Italy
| | - Francesco Rossi
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Via Costantinopoli 16, 80138 Naples, Italy
| | - Antonella De Angelis
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Via Costantinopoli 16, 80138 Naples, Italy.
| | - Konrad Urbanek
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Via Costantinopoli 16, 80138 Naples, Italy
| |
Collapse
|
169
|
Koh JS, Yi CO, Heo RW, Ahn JW, Park JR, Lee JE, Kim JH, Hwang JY, Roh GS. Protective effect of cilostazol against doxorubicin-induced cardiomyopathy in mice. Free Radic Biol Med 2015; 89:54-61. [PMID: 26191652 DOI: 10.1016/j.freeradbiomed.2015.07.016] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 07/10/2015] [Accepted: 07/15/2015] [Indexed: 10/23/2022]
Abstract
Doxorubicin (Dox) is an effective anti-cancer drug, but its use is limited because of its adverse effect of inducing irreversible dilated cardiomyopathy. Cilostazol (Cilo), a potent phosphodiesterase III inhibitor, has been reported to have an anti-inflammatory effect. Here, we investigated whether Cilo has a protective effect against Dox-induced cardiomyopathy (DIC). Mice were randomly divided into four groups: saline control, Dox (15 mg/kg), Dox (15 mg/kg) plus Cilo (50mg/kg), and Cilo (50mg/kg). The results showed that the coadministration of Dox and Cilo significantly enhanced left-ventricular systolic function compared with Dox alone. In addition, Cilo treatment significantly reduced Dox-induced perivascular fibrosis, collagen concentration, and connective growth factor expression in the heart. Also, Cilo administration markedly reduced Dox-induced levels of serum B-type natriuretic peptide, dysferlin, high-mobility group protein B1, Toll-like receptor 4, nuclear factor-κB p65, and cyclooxygenase-2. Furthermore, Cilo treatment significantly reduced Dox-induced oxidative stress by lowering the translocation of Nrf2 into the nucleus and the expression of NQO1, heme oxygenase 1, and superoxide dismutase-1. Our results suggest that Cilo may be a potential antifibrotic, antioxidative, and anti-inflammatory drug for DIC.
Collapse
Affiliation(s)
- Jin Sin Koh
- Division of Cardiology, Department of Internal Medicine, Gyeongsang National University Hospital, Gyeongsang National University School of Medicine, Jinju 660-751, Republic of Korea
| | - Chin-ok Yi
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju 660-751, Republic of Korea
| | - Rok Won Heo
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju 660-751, Republic of Korea
| | - Jong-Wha Ahn
- Division of Cardiology, Department of Internal Medicine, Gyeongsang National University Hospital, Gyeongsang National University School of Medicine, Jinju 660-751, Republic of Korea
| | - Jeong Rang Park
- Division of Cardiology, Department of Internal Medicine, Gyeongsang National University Hospital, Gyeongsang National University School of Medicine, Jinju 660-751, Republic of Korea
| | - Jung Eun Lee
- Department of Thoracic and Cardiovascular Surgery, Gyeongsang National University Hospital, Gyeongsang National University School of Medicine, Jinju 660-751, Republic of Korea
| | - Jung-Hwan Kim
- Department of Pharmacology, Institute of Health Sciences, Gyeongsang National University School of Medicine,Jinju 660-751, Republic of Korea
| | - Jin-Yong Hwang
- Division of Cardiology, Department of Internal Medicine, Gyeongsang National University Hospital, Gyeongsang National University School of Medicine, Jinju 660-751, Republic of Korea.
| | - Gu Seob Roh
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju 660-751, Republic of Korea.
| |
Collapse
|
170
|
Interaction of Hydrogen Sulfide with Nitric Oxide in the Cardiovascular System. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:6904327. [PMID: 26640616 PMCID: PMC4657111 DOI: 10.1155/2016/6904327] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2015] [Accepted: 06/21/2015] [Indexed: 01/07/2023]
Abstract
Historically acknowledged as toxic gases, hydrogen sulfide (H2S) and nitric oxide (NO) are now recognized as the predominant members of a new family of signaling molecules, “gasotransmitters” in mammals. While H2S is biosynthesized by three constitutively expressed enzymes (CBS, CSE, and 3-MST) from L-cysteine and homocysteine, NO is generated endogenously from L-arginine by the action of various isoforms of NOS. Both gases have been transpired as the key and independent regulators of many physiological functions in mammalian cardiovascular, nervous, gastrointestinal, respiratory, and immune systems. The analogy between these two gasotransmitters is evident not only from their paracrine mode of signaling, but also from the identical and/or shared signaling transduction pathways. With the plethora of research in the pathophysiological role of gasotransmitters in various systems, the existence of interplay between these gases is being widely accepted. Chemical interaction between NO and H2S may generate nitroxyl (HNO), which plays a specific effective role within the cardiovascular system. In this review article, we have attempted to provide current understanding of the individual and interactive roles of H2S and NO signaling in mammalian cardiovascular system, focusing particularly on heart contractility, cardioprotection, vascular tone, angiogenesis, and oxidative stress.
Collapse
|
171
|
Varga ZV, Ferdinandy P, Liaudet L, Pacher P. Drug-induced mitochondrial dysfunction and cardiotoxicity. Am J Physiol Heart Circ Physiol 2015; 309:H1453-H1467. [PMID: 26386112 PMCID: PMC4666974 DOI: 10.1152/ajpheart.00554.2015] [Citation(s) in RCA: 349] [Impact Index Per Article: 34.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 09/15/2015] [Indexed: 12/14/2022]
Abstract
Mitochondria has an essential role in myocardial tissue homeostasis; thus deterioration in mitochondrial function eventually leads to cardiomyocyte and endothelial cell death and consequent cardiovascular dysfunction. Several chemical compounds and drugs have been known to directly or indirectly modulate cardiac mitochondrial function, which can account both for the toxicological and pharmacological properties of these substances. In many cases, toxicity problems appear only in the presence of additional cardiovascular disease conditions or develop months/years following the exposure, making the diagnosis difficult. Cardiotoxic agents affecting mitochondria include several widely used anticancer drugs [anthracyclines (Doxorubicin/Adriamycin), cisplatin, trastuzumab (Herceptin), arsenic trioxide (Trisenox), mitoxantrone (Novantrone), imatinib (Gleevec), bevacizumab (Avastin), sunitinib (Sutent), and sorafenib (Nevaxar)], antiviral compound azidothymidine (AZT, Zidovudine) and several oral antidiabetics [e.g., rosiglitazone (Avandia)]. Illicit drugs such as alcohol, cocaine, methamphetamine, ecstasy, and synthetic cannabinoids (spice, K2) may also induce mitochondria-related cardiotoxicity. Mitochondrial toxicity develops due to various mechanisms involving interference with the mitochondrial respiratory chain (e.g., uncoupling) or inhibition of the important mitochondrial enzymes (oxidative phosphorylation, Szent-Györgyi-Krebs cycle, mitochondrial DNA replication, ADP/ATP translocator). The final phase of mitochondrial dysfunction induces loss of mitochondrial membrane potential and an increase in mitochondrial oxidative/nitrative stress, eventually culminating into cell death. This review aims to discuss the mechanisms of mitochondrion-mediated cardiotoxicity of commonly used drugs and some potential cardioprotective strategies to prevent these toxicities.
Collapse
Affiliation(s)
- Zoltán V Varga
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institutes of Health/National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland; Cardiometabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Peter Ferdinandy
- Cardiometabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Pharmahungary Group, Szeged, Hungary; and
| | - Lucas Liaudet
- Department of Intensive Care Medicine BH 08-621-University Hospital Medical Center, Lausanne, Switzerland
| | - Pál Pacher
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institutes of Health/National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland;
| |
Collapse
|
172
|
Efremova AS, Shram SI, Myasoedov NF. Doxorubicin causes transient activation of protein poly(ADP-ribosyl)ation in H9c2 cardiomyocytes. DOKL BIOCHEM BIOPHYS 2015; 464:333-7. [PMID: 26518562 DOI: 10.1134/s1607672915050178] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Indexed: 11/22/2022]
Abstract
Possible involvement of the system of protein poly(ADP-ribosyl)ation in the mechanisms of cardiotoxicity of doxorubicin, one of the most frequently used anticancer drug, was studied in cultures of cardiomyocytes H9c2. The treatment of H9c2 cells with doxorubicin (1 µM) led to a transient (after 6 h of incubation) increase in the nuclear level of poly(ADP-ribosyl)ated proteins. The observed data indirectly indicate the development of genotoxic stress in the doxorubicin-treated cells, probably caused by the stimulatory effects of doxorubicin and its metabolites on the production of reactive oxygen and nitrogen species.
Collapse
Affiliation(s)
- A S Efremova
- Institute of Molecular Genetics, Russian Academy of Sciences, pl. Akademika Kurchatova 46, Moscow, 123182, Russia
| | - S I Shram
- Institute of Molecular Genetics, Russian Academy of Sciences, pl. Akademika Kurchatova 46, Moscow, 123182, Russia.
| | - N F Myasoedov
- Institute of Molecular Genetics, Russian Academy of Sciences, pl. Akademika Kurchatova 46, Moscow, 123182, Russia
| |
Collapse
|
173
|
Kwatra M, Kumar V, Jangra A, Mishra M, Ahmed S, Ghosh P, Vohora D, Khanam R. Ameliorative effect of naringin against doxorubicin-induced acute cardiac toxicity in rats. PHARMACEUTICAL BIOLOGY 2015; 54:637-647. [PMID: 26471226 DOI: 10.3109/13880209.2015.1070879] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
CONTEXT Doxorubicin (Dox) is one of the most active chemotherapeutic agents used to treat various types of cancers. Its clinical utility is compromised due to fatal cardiac toxicity characterized by an irreversible cardiomyopathy. OBJECTIVE This study evaluates the cardioprotective potential of naringin (NR) against Dox-induced acute cardiac toxicity in rats. MATERIALS AND METHODS Male Wistar rats were randomly divided into five groups. NR (50 and 100 mg/kg) was administered intraperitoneally (i.p.) daily from 0 to 14 d. Doxorubicin (15 mg/kg, i.p.) was given as a single dose on the 10th day. On the 14th day, all animals were sacrificed and oxidative stress parameters that include malondialdehyde (MDA), glutathione (GSH) level, superoxide dismutase (SOD), catalase (CAT) activities, and all mitochondrial complexes (I-IV) activities were evaluated along with histopathological studies of the heart. RESULTS Doxorubicin-induced cardiotoxicity was confirmed by increased (p < 0.05) MDA, decreased (p < 0.05) GSH levels, SOD, and CAT activities, mitochondrial complexes (I-IV) activities in the heart tissue. NR (100 mg/kg) showed cardioprotection as evident from significant decreased MDA (p < 0.001) level, raised (p < 0.001) GSH level, SOD and CAT activities and increased mitochondrial complexes I (p < 0.01), II (p < 0.001), III (p < 0.001), and IV (p < 0.05) activities. Further, Dox-induced cardiotoxicity was confirmed by histopathological studies. These obtained results indicated the protective role of NR against Dox-induced cardiac toxicity in rats. CONCLUSION NR can be used in combination with Dox due to its high cardioprotective effect against Dox-induced cardiomyopathy.
Collapse
Affiliation(s)
- Mohit Kwatra
- a Pharmacology Research Laboratory, Department of Pharmacology, Faculty of Pharmacy, Jamia Hamdard (Hamdard University) , New Delhi , India
| | - Vikas Kumar
- a Pharmacology Research Laboratory, Department of Pharmacology, Faculty of Pharmacy, Jamia Hamdard (Hamdard University) , New Delhi , India
| | - Ashok Jangra
- b Department of Pharmacology and Toxicology , National Institute of Pharmaceutical Education and Research Guwahati , Guwahati , Assam , India
| | - Murli Mishra
- c Department of Toxicology and Cancer Biology , College of Medicine, University of Kentucky , Lexington , KY , USA
| | - Sahabuddin Ahmed
- b Department of Pharmacology and Toxicology , National Institute of Pharmaceutical Education and Research Guwahati , Guwahati , Assam , India
| | - Pinaki Ghosh
- d Department of Pharmacology , Bharati Vidyapeeth University, Poona College of Pharmacy , Erandwane , Pune , Maharashtra , India , and
| | - Divya Vohora
- a Pharmacology Research Laboratory, Department of Pharmacology, Faculty of Pharmacy, Jamia Hamdard (Hamdard University) , New Delhi , India
| | - Razia Khanam
- a Pharmacology Research Laboratory, Department of Pharmacology, Faculty of Pharmacy, Jamia Hamdard (Hamdard University) , New Delhi , India
- e Department of Pharmacology , Gulf Medical University , Ajman , United Arab Emirates
| |
Collapse
|
174
|
Angsutararux P, Luanpitpong S, Issaragrisil S. Chemotherapy-Induced Cardiotoxicity: Overview of the Roles of Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:795602. [PMID: 26491536 PMCID: PMC4602327 DOI: 10.1155/2015/795602] [Citation(s) in RCA: 181] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 05/17/2015] [Indexed: 02/02/2023]
Abstract
Chemotherapy-induced cardiotoxicity is a serious complication that poses a serious threat to life and limits the clinical use of various chemotherapeutic agents, particularly the anthracyclines. Understanding molecular mechanisms of chemotherapy-induced cardiotoxicity is a key to effective preventive strategies and improved chemotherapy regimen. Although no reliable and effective preventive treatment has become available, numerous evidence demonstrates that chemotherapy-induced cardiotoxicity involves the generation of reactive oxygen species (ROS). This review provides an overview of the roles of oxidative stress in chemotherapy-induced cardiotoxicity using doxorubicin, which is one of the most effective chemotherapeutic agents against a wide range of cancers, as an example. Current understanding in the molecular mechanisms of ROS-mediated cardiotoxicity will be explored and discussed, with emphasis on cardiomyocyte apoptosis leading to cardiomyopathy. The review will conclude with perspectives on model development needed to facilitate further progress and understanding on chemotherapy-induced cardiotoxicity.
Collapse
Affiliation(s)
- Paweorn Angsutararux
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Sudjit Luanpitpong
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Surapol Issaragrisil
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| |
Collapse
|
175
|
Tocchetti CG, Carpi A, Coppola C, Quintavalle C, Rea D, Campesan M, Arcari A, Piscopo G, Cipresso C, Monti MG, De Lorenzo C, Arra C, Condorelli G, Di Lisa F, Maurea N. Ranolazine protects from doxorubicin-induced oxidative stress and cardiac dysfunction. Eur J Heart Fail 2015; 16:358-66. [PMID: 24464789 DOI: 10.1002/ejhf.50] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 11/18/2013] [Accepted: 11/22/2013] [Indexed: 12/18/2022] Open
Abstract
AIMS Doxorubicin is widely used against cancer; however, it can produce heart failure (HF). Among other hallmarks, oxidative stress is a major contributor to HF pathophysiology. The late INa inhibitor ranolazine has proven effective in treating experimental HF. Since elevated [Na+]i is present in failing myocytes, and has been recently linked with reactive oxygen species (ROS) production, our aim was to assess whether ranolazine prevents doxorubicin-induced cardiotoxicity, and whether blunted oxidative stress is a mechanism accounting for such protection. METHODS AND RESULT In C57BL6 mice, doxorubicin treatment for 7 days produced LV dilation and decreased echo-measured fractional shortening (FS). Ranolazine (305 mg/kg/day) prevented LV dilation and dysfunction when co-administered with doxorubicin. Doxorubicin-induced cardiotoxicity was accompanied instead by elevations in atrial natriuretic peptide (ANP), BNP, connective tissue growth factor (CTGF), and matrix metalloproteinase 2 (MMP2) mRNAs, which were not elevated on co-treatment with ranolazine. Alterations in extracellular matrix remodelling were confirmed by an increase in interstitial collagen, which did not rise in ranolazine-co-treated hearts. Levels of poly(ADP-ribose) polymerase (PARP) and pro-caspase-3 measured by western blotting were lowered with doxorubicin, with increased cleavage of caspase-3, indicating activation of the proapoptotic machinery. Again, ranolazine prevented this activation. Furthermore, in HL-1 cardiomyocytes transfected with HyPer to monitor H2O2 emission, besides reducing the extent of cell death, ranolazine prevented the occurrence of oxidative stress caused by doxorubicin. Interestingly, similar protective results were obtained with the Na+/Ca2+ exchanger (NCX) inhibitor KB-R7943. CONCLUSIONS Ranolazine protects against experimental doxorubicin cardiotoxicity. Such protection is accompanied by a reduction in oxidative stress, suggesting that INa modulates cardiac redox balance, resulting in functional and morphological derangements.
Collapse
|
176
|
Mansour HH, El Kiki SM, Hasan HF. Protective effect of N-acetylcysteine on cyclophosphamide-induced cardiotoxicity in rats. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2015; 40:417-422. [PMID: 26262887 DOI: 10.1016/j.etap.2015.07.013] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Revised: 07/14/2015] [Accepted: 07/18/2015] [Indexed: 06/04/2023]
Abstract
Cyclophosphamide (CP) is an oxazaphosphorine nitrogen mustard alkylating drug used for the treatment of chronic and acute leukemias, lymphoma, myeloma, and cancers of the breast and ovary. It is known to cause severe cardiac toxicity. This study investigated the protective effect of N-Acetylcysteine (NAC) on CP-induced cardiotoxicity in rats. CP resulted in a significant increase in serum aminotransferases, creatine kinase (CK), lactate dehydrogenase(LDH) enzymes, asymmetric dimethylarginine and tumor necrosis factor-α and significant decrease in total nitrate/nitrite(NOx). In cardiac tissues, a single dose of CP (200mg/kg, i.p.) resulted in significant increase in malondialdehyde and NOx and a significant decrease in reduced glutathione content, glutathione peroxidase, catalase, and superoxide dismutase activities. Interestingly, Administration of NAC (200mg/kg, i.p.) for 5 days prior to CP attenuates all the biochemical changes induced by CP. These results revealed that NAC attenuates CP-induced cardiotoxicity by inhibiting oxidative and nitrosative stress and preserving the activity of antioxidant enzymes.
Collapse
Affiliation(s)
- Heba H Mansour
- Health Radiation Research Department, National Center for Radiation Research and Technology, PO Box 29, Nasr City, Cairo, Egypt.
| | - Shereen M El Kiki
- Health Radiation Research Department, National Center for Radiation Research and Technology, PO Box 29, Nasr City, Cairo, Egypt
| | - Hesham F Hasan
- Radiation Biology Department, National Center for Radiation Research and Technology, Atomic Energy Authority, Cairo, Egypt
| |
Collapse
|
177
|
Kwon HK, Shin HJ, Lee JH, Park SH, Kwon MC, Panneerselvam S, Lee CG, Kim SG, Kim JH, Choi S. Etoposide Induces Necrosis Through p53-Mediated Antiapoptosis in Human Kidney Proximal Tubule Cells. Toxicol Sci 2015; 148:204-19. [PMID: 26259609 DOI: 10.1093/toxsci/kfv182] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The p53 protein is an important transcription factor that modulates signaling pathways for both cell death and survival. Its antiapoptotic mechanisms that correlate with necrotic and apoptotic cell death are not well understood. Here, we report that etoposide promotes progression of the DNA damage response as well as necrotic morphological changes including plasma membrane rupture using carbon nanotube-tipped/atomic force microscopy (CNT/AFM) probes in human kidney proximal tubule (HK-2) cells. Inhibition of p53 abrogated cell cycle arrest and led to a decrease in the expression levels of repair proteins that were induced by DNA damage. Mitochondrial biogenesis and cytosolic production of reactive oxygen species were also reduced after p53 inhibition; the latter change induced mitochondrial superoxide accumulation and mitochondrial damage, which triggered the activation of caspase 3. Inhibition of p53 also led to a loss of cell adhesion and converted necrotic cell death to apoptotic cell death, with appreciable cell shrinkage and appearance of apoptotic bodies that were observed using CNT/AFM probes. Thus, our study demonstrated that p53 protects against apoptosis, and leads to etoposide-induced necrosis. These results are expected to aid in the understanding of mechanism of antiapoptosis and its relationship to cell death.
Collapse
Affiliation(s)
- Hyuk-Kwon Kwon
- *Department of Molecular Science and Technology, Ajou University, Suwon 443-749, Korea and
| | - Hyeon-Jun Shin
- *Department of Molecular Science and Technology, Ajou University, Suwon 443-749, Korea and
| | - Jae-Hyeok Lee
- *Department of Molecular Science and Technology, Ajou University, Suwon 443-749, Korea and
| | - Seol-Hee Park
- *Department of Molecular Science and Technology, Ajou University, Suwon 443-749, Korea and
| | - Min-Cheol Kwon
- *Department of Molecular Science and Technology, Ajou University, Suwon 443-749, Korea and
| | - Suresh Panneerselvam
- *Department of Molecular Science and Technology, Ajou University, Suwon 443-749, Korea and
| | - Chan Gyu Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 151-742, Korea
| | - Sang Geon Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 151-742, Korea
| | - Jae-Ho Kim
- *Department of Molecular Science and Technology, Ajou University, Suwon 443-749, Korea and
| | - Sangdun Choi
- *Department of Molecular Science and Technology, Ajou University, Suwon 443-749, Korea and
| |
Collapse
|
178
|
Yao CX, Shi JC, Ma CX, Xiong CJ, Song YL, Zhang SF, Zhang SF, Zang MX, Xue LX. EGF Protects Cells Against Dox-Induced Growth Arrest Through Activating Cyclin D1 Expression. J Cell Biochem 2015; 116:1755-65. [DOI: 10.1002/jcb.25134] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 02/06/2015] [Indexed: 02/01/2023]
Affiliation(s)
- Chun-Xia Yao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences; Zhengzhou University; Zhengzhou City Henan 450001 China
| | - Jia-Chen Shi
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences; Zhengzhou University; Zhengzhou City Henan 450001 China
| | - Cai-Xia Ma
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences; Zhengzhou University; Zhengzhou City Henan 450001 China
| | - Cheng-Juan Xiong
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences; Zhengzhou University; Zhengzhou City Henan 450001 China
| | - Yang-Liu Song
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences; Zhengzhou University; Zhengzhou City Henan 450001 China
| | - Shu-Feng Zhang
- The People's Hospital of Henan Province; Zhengzhou University; Zhengzhou Henan 450001 China
| | - Shan-Feng Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences; Zhengzhou University; Zhengzhou City Henan 450001 China
| | - Ming-Xi Zang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences; Zhengzhou University; Zhengzhou City Henan 450001 China
| | - Li-Xiang Xue
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences; Peking University; Beijing 100191 China
| |
Collapse
|
179
|
High Throughput Screening Identifies a Novel Compound Protecting Cardiomyocytes from Doxorubicin-Induced Damage. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:178513. [PMID: 26137186 PMCID: PMC4475553 DOI: 10.1155/2015/178513] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 04/27/2015] [Accepted: 05/20/2015] [Indexed: 12/01/2022]
Abstract
Antracyclines are effective antitumor agents. One of the most commonly used antracyclines is doxorubicin, which can be successfully used to treat a diverse spectrum of tumors. Application of these drugs is limited by their cardiotoxic effect, which is determined by a lifetime cumulative dose. We set out to identify by high throughput screening cardioprotective compounds protecting cardiomyocytes from doxorubicin-induced injury. Ten thousand compounds of ChemBridge's DIVERSet compound library were screened to identify compounds that can protect H9C2 rat cardiomyocytes against doxorubicin-induced cell death. The most effective compound proved protective in doxorubicin-treated primary rat cardiomyocytes and was further characterized to demonstrate that it significantly decreased doxorubicin-induced apoptotic and necrotic cell death and inhibited doxorubicin-induced activation of JNK MAP kinase without having considerable radical scavenging effect or interfering with the antitumor effect of doxorubicin. In fact the compound identified as 3-[2-(4-ethylphenyl)-2-oxoethyl]-1,2-dimethyl-1H-3,1-benzimidazol-3-ium bromide was toxic to all tumor cell lines tested even without doxorubicine treatment. This benzimidazole compound may lead, through further optimalization, to the development of a drug candidate protecting the heart from doxorubicin-induced injury.
Collapse
|
180
|
Yamaguchi SI, Ueki A, Sugihara E, Onishi N, Yaguchi T, Kawakami Y, Horiuchi K, Morioka H, Matsumoto M, Nakamura M, Muto A, Toyama Y, Saya H, Shimizu T. Synergistic antiproliferative effect of imatinib and adriamycin in platelet-derived growth factor receptor-expressing osteosarcoma cells. Cancer Sci 2015; 106:875-82. [PMID: 25940371 PMCID: PMC4520639 DOI: 10.1111/cas.12686] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 04/23/2015] [Accepted: 04/27/2015] [Indexed: 12/20/2022] Open
Abstract
Osteosarcoma (OS) is the most frequent primary solid malignant tumor of bone. Its prognosis remains poor in the substantial proportion of patients who do not respond to chemotherapy and novel therapeutic options are therefore needed. We previously established a mouse model that mimics the aggressive behavior of human OS. Enzyme-linked immunosorbent assay-based screening of such mouse tumor lysates identified platelet-derived growth factor–BB (PDGF-BB) as an abundant soluble factor, the gene for which was expressed dominantly in surrounding non-malignant cells of the tumor, whereas that for the cognate receptor (PDGF receptor β) was highly expressed in OS cells. Platelet-derived growth factor-BB induced activation of both MEK–ERK and phosphatidylinositol 3-kinase–protein kinase B signaling pathways and promoted survival in OS cells deprived of serum, and these effects were blocked by the PDGF receptor inhibitor imatinib. However, these actions of PDGF-BB and imatinib were mostly masked in the presence of serum. Whereas imatinib alone did not manifest an antitumor effect in mice harboring OS tumors, combined treatment with imatinib and adriamycin exerted a synergistic antiproliferative effect on OS cells in vivo. These results suggest that treatment of OS with imatinib is effective only when cell survival is dependent on PDGF signaling or when imatinib is combined with another therapeutic intervention that renders the tumor cells susceptible to imatinib action, such as by inducing cellular stress.
Collapse
Affiliation(s)
- Sayaka I Yamaguchi
- Division of Gene Regulation, Institute for Advanced Medical Research (IAMR), Keio University School of Medicine, Tokyo, Japan.,Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Arisa Ueki
- Division of Gene Regulation, Institute for Advanced Medical Research (IAMR), Keio University School of Medicine, Tokyo, Japan
| | - Eiji Sugihara
- Division of Gene Regulation, Institute for Advanced Medical Research (IAMR), Keio University School of Medicine, Tokyo, Japan.,Core research for evolutionary science and technology (CREST), Japan Science and Technology Agency, Tokyo, Japan
| | - Nobuyuki Onishi
- Division of Gene Regulation, Institute for Advanced Medical Research (IAMR), Keio University School of Medicine, Tokyo, Japan
| | - Tomonori Yaguchi
- Division of Cellular Signaling, Institute for Advanced Medical Research (IAMR), Keio University School of Medicine, Tokyo, Japan
| | - Yutaka Kawakami
- Division of Cellular Signaling, Institute for Advanced Medical Research (IAMR), Keio University School of Medicine, Tokyo, Japan
| | - Keisuke Horiuchi
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Hideo Morioka
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Morio Matsumoto
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Masaya Nakamura
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Akihiro Muto
- Department of Pathophysiology, Hoshi University, Tokyo, Japan
| | - Yoshiaki Toyama
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Hideyuki Saya
- Division of Gene Regulation, Institute for Advanced Medical Research (IAMR), Keio University School of Medicine, Tokyo, Japan.,Core research for evolutionary science and technology (CREST), Japan Science and Technology Agency, Tokyo, Japan
| | - Takatsune Shimizu
- Division of Gene Regulation, Institute for Advanced Medical Research (IAMR), Keio University School of Medicine, Tokyo, Japan.,Core research for evolutionary science and technology (CREST), Japan Science and Technology Agency, Tokyo, Japan.,Department of Pathophysiology, Hoshi University, Tokyo, Japan
| |
Collapse
|
181
|
Wang HL, Gao JP, Han YL, Xu X, Wu R, Gao Y, Cui XH. Comparative studies of polydatin and resveratrol on mutual transformation and antioxidative effect in vivo. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2015; 22:553-559. [PMID: 25981921 DOI: 10.1016/j.phymed.2015.03.014] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2014] [Revised: 03/06/2015] [Accepted: 03/24/2015] [Indexed: 06/04/2023]
Abstract
BACKGROUND Polydatin and resveratrol are extractives of radix or rhizoma of Polygonum cuspidatum, and as the glycoside forms, it is a natural precursor of resveratrol. PURPOSE In this study, we aimed to explore the mutual transformation between polydatin and resveratrol in rats, and to compare the antioxidative effect of them in vivo. STUDY DESIGN In this study, we analyzed the serum molar concentration of polydatin and resveratrol after oral administration in rats and evaluated the anti-oxidative stress effects of them using a mouse model. METHODS Rats were orally administered polydatin or resveratrol and the concentration of them in serum were analyzed by high performance liquid chromatography (HPLC). Their antioxidative effect was compared in mice with oxidative stress cardiomyopathy induced by doxorubicin (DOX). RESULTS The results showed that polydatin and resveratrol could mutually transform in vivo, the molar concentration of polydatin in serum was always averagely 3.35 and 4.28 times as much as resveratrol after oral administration of polydatin and resveratrol at 200 mg/kg, respectively. Both polydatin and resveratrol could significantly decrease the content of malonydialdehyde (MDA), promote the activities of total superoxide dismutase (T-SOD), catalase (CAT) and glutathione peroxidase (GSH-Px) in plasma, and increase the content of glutathione (GSH) in myocardial tissue. The effect of polydatin surpassed resveratrol, particularly embodied in increasing the activities of T-SOD and CAT, and the content of GSH. CONCLUSION It illustrates that polydatin is the main substance in serum after intragastric administration with polydatin or resveratrol, and the mutual transformation between polydatin and resveratrol keeps balance; they both have the ability of antioxidative stress in vivo, and polydatin has a better effect than resveratrol, which hints that polydatin may be a substitute for resveratrol in antioxidant for clinical use.
Collapse
Affiliation(s)
- Hui-Lin Wang
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China.
| | - Jian-Ping Gao
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China.
| | - Yu-Liang Han
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai 201418, China.
| | - Xu Xu
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai 201418, China.
| | - Rong Wu
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China.
| | - Yan Gao
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China.
| | - Xiao-Hua Cui
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China.
| |
Collapse
|
182
|
Chu W, Chepetan A, Zhou D, Shoghi KI, Xu J, Dugan LL, Gropler RJ, Mintun MA, Mach RH. Development of a PET radiotracer for non-invasive imaging of the reactive oxygen species, superoxide, in vivo. Org Biomol Chem 2015; 12:4421-31. [PMID: 24847866 DOI: 10.1039/c3ob42379d] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Reactive oxygen species (ROS) have been implicated in the pathogenesis of a wide range of human disease states and drug toxicities, but development of imaging tools to study ROS biology in vivo remains a challenge. Here we synthesized and validated a novel PET tracer (12) and its (18)F radiolabeled version [(18)F]12 to allow PET ( positron emission tomography) imaging of superoxide in vivo. Initial analysis of ROS reaction kinetics found that compound 12 was rapidly and selectively oxidized by superoxide, but not other ROS. Cell culture studies in EMT6 cells exposed to the cancer chemotherapeutic agent Doxorubicin (DOX), which activates the superoxide-generating enzyme, NADPH oxidase, showed that compound 12 was a sensitive and specific probe for superoxide in cells. The microPET imaging of heart in mice with DOX-induced cardiac inflammation observed 2-fold greater oxidation of [(18)F]12 in the DOX-treated mice compared to controls (p = 0.02), the results were confirmed by distribution studies on organs subsequently removed from the mice and HPLC analysis of [(18)F] radioactivity compounds. These data indicate that compound 12 is a useful PET tracer to imaging ROS in vivo.
Collapse
Affiliation(s)
- Wenhua Chu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
183
|
Roca-Alonso L, Castellano L, Mills A, Dabrowska AF, Sikkel MB, Pellegrino L, Jacob J, Frampton AE, Krell J, Coombes RC, Harding SE, Lyon AR, Stebbing J. Myocardial MiR-30 downregulation triggered by doxorubicin drives alterations in β-adrenergic signaling and enhances apoptosis. Cell Death Dis 2015; 6:e1754. [PMID: 25950484 PMCID: PMC4669718 DOI: 10.1038/cddis.2015.89] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 12/14/2014] [Accepted: 01/12/2015] [Indexed: 12/22/2022]
Abstract
The use of anthracyclines such as doxorubicin (DOX) has improved outcome in cancer patients, yet associated risks of cardiomyopathy have limited their clinical application. DOX-associated cardiotoxicity is frequently irreversible and typically progresses to heart failure (HF) but our understanding of molecular mechanisms underlying this and essential for development of cardioprotective strategies remains largely obscure. As microRNAs (miRNAs) have been shown to play potent regulatory roles in both cardiovascular disease and cancer, we investigated miRNA changes in DOX-induced HF and the alteration of cellular processes downstream. Myocardial miRNA profiling was performed after DOX-induced injury, either via acute application to isolated cardiomyocytes or via chronic exposure in vivo, and compared with miRNA profiles from remodeled hearts following myocardial infarction. The miR-30 family was downregulated in all three models. We describe here that miR-30 act regulating the β-adrenergic pathway, where preferential β1- and β2-adrenoceptor (β1AR and β2AR) direct inhibition is combined with Giα-2 targeting for fine-tuning. Importantly, we show that miR-30 also target the pro-apoptotic gene BNIP3L/NIX. In aggregate, we demonstrate that high miR-30 levels are protective against DOX toxicity and correlate this in turn with lower reactive oxygen species generation. In addition, we identify GATA-6 as a mediator of DOX-associated reductions in miR-30 expression. In conclusion, we describe that DOX causes acute and sustained miR-30 downregulation in cardiomyocytes via GATA-6. miR-30 overexpression protects cardiac cells from DOX-induced apoptosis, and its maintenance represents a potential cardioprotective and anti-tumorigenic strategy for anthracyclines.
Collapse
Affiliation(s)
- L Roca-Alonso
- Division of Oncology, Department of Surgery and Cancer, 1st Floor, Imperial Centre for Translational and Experimental Medicine (ICTEM), Imperial College, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | - L Castellano
- Division of Oncology, Department of Surgery and Cancer, 1st Floor, Imperial Centre for Translational and Experimental Medicine (ICTEM), Imperial College, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | - A Mills
- National Heart and Lung Institute, Imperial College, 4th Floor, ICTEM, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | - A F Dabrowska
- Division of Oncology, Department of Surgery and Cancer, 1st Floor, Imperial Centre for Translational and Experimental Medicine (ICTEM), Imperial College, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | - M B Sikkel
- National Heart and Lung Institute, Imperial College, 4th Floor, ICTEM, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | - L Pellegrino
- Division of Oncology, Department of Surgery and Cancer, 1st Floor, Imperial Centre for Translational and Experimental Medicine (ICTEM), Imperial College, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | - J Jacob
- Division of Oncology, Department of Surgery and Cancer, 1st Floor, Imperial Centre for Translational and Experimental Medicine (ICTEM), Imperial College, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | - A E Frampton
- Division of Oncology, Department of Surgery and Cancer, 1st Floor, Imperial Centre for Translational and Experimental Medicine (ICTEM), Imperial College, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
- Hepato-Pancreato-Biliary Surgical Unit, Department of Surgery and Cancer, Imperial College, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | - J Krell
- Division of Oncology, Department of Surgery and Cancer, 1st Floor, Imperial Centre for Translational and Experimental Medicine (ICTEM), Imperial College, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | - R C Coombes
- Division of Oncology, Department of Surgery and Cancer, 1st Floor, Imperial Centre for Translational and Experimental Medicine (ICTEM), Imperial College, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | - S E Harding
- National Heart and Lung Institute, Imperial College, 4th Floor, ICTEM, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | - A R Lyon
- National Heart and Lung Institute, Imperial College, 4th Floor, ICTEM, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
- NIHR Cardiovascular Biomedical Research Unit, Royal Brompton Hospital, Imperial college, London SW3 6NP, UK
| | - J Stebbing
- Division of Oncology, Department of Surgery and Cancer, 1st Floor, Imperial Centre for Translational and Experimental Medicine (ICTEM), Imperial College, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| |
Collapse
|
184
|
Li B, Kim DS, Yadav RK, Kim HR, Chae HJ. Sulforaphane prevents doxorubicin-induced oxidative stress and cell death in rat H9c2 cells. Int J Mol Med 2015; 36:53-64. [PMID: 25936432 PMCID: PMC4494600 DOI: 10.3892/ijmm.2015.2199] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 04/22/2015] [Indexed: 12/18/2022] Open
Abstract
Sulforaphane, a natural isothiocyanate compound found in cruciferous vegetables, has been shown to exert cardioprotective effects during ischemic heart injury. However, the effects of sulforaphane on cardiotoxicity induced by doxorubicin are unknown. Thus, in the present study, H9c2 rat myoblasts were pre-treated with sulforaphane and its effects on cardiotoxicity were then examined. The results revealed that the pre-treatment of H9c2 rat myoblasts with sulforaphane decreased the apoptotic cell number (as shown by trypan blue exclusion assay) and the expression of pro-apoptotic proteins (Bax, caspase-3 and cytochrome c; as shown by western blot analysis and immunostaining), as well as the doxorubicin-induced increase in mitochondrial membrane potential (measured by JC-1 assay). Furthermore, sulforaphane increased the mRNA and protein expression of heme oxygenase-1 (HO-1, measured by RT-qPCR), which consequently reduced the levels of reactive oxygen species (ROS, measured using MitoSOX Red reagent) in the mitochondria which were induced by doxorubicin. The cardioprotective effects of sulforaphane were found to be mediated by the activation of the Kelch-like ECH-associated protein 1 (Keap1)/NF-E2-related factor-2 (Nrf2)/antioxidant-responsive element (ARE) pathway, which in turn mediates the induction of HO-1. Taken together, the findings of this study demonstrate that sulforaphane prevents doxorubicin-induced oxidative stress and cell death in H9c2 cells through the induction of HO-1 expression.
Collapse
Affiliation(s)
- Bo Li
- Department of Pharmacology and Institute of Cardiovascular Research, School of Medicine, Chonbuk National University, Jeonju, Chonbuk 561-180, Republic of Korea
| | - Do Sung Kim
- Department of Pharmacology and Institute of Cardiovascular Research, School of Medicine, Chonbuk National University, Jeonju, Chonbuk 561-180, Republic of Korea
| | - Raj Kumar Yadav
- Department of Pharmacology and Institute of Cardiovascular Research, School of Medicine, Chonbuk National University, Jeonju, Chonbuk 561-180, Republic of Korea
| | - Hyung Ryong Kim
- Department of Dental Pharmacology and Wonkwang Biomaterial Implant Research Institute, School of Dentistry, Wonkwang University, Iksan, Chonbuk 570-749, Republic of Korea
| | - Han Jung Chae
- Department of Pharmacology and Institute of Cardiovascular Research, School of Medicine, Chonbuk National University, Jeonju, Chonbuk 561-180, Republic of Korea
| |
Collapse
|
185
|
Theodorakis N, Maluccio M, Skill N. Murine study of portal hypertension associated endothelin-1 hypo-response. World J Gastroenterol 2015; 21:4817-4828. [PMID: 25944995 PMCID: PMC4408454 DOI: 10.3748/wjg.v21.i16.4817] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 11/03/2014] [Accepted: 12/08/2014] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate endothelin-1 hypo-responsive associated with portal hypertension in order to improve patient treatment outcomes.
METHODS: Wild type, eNOS-/- and iNOS-/- mice received partial portal vein ligation surgery to induce portal hypertension or sham surgery. Development of portal hypertension was determined by measuring the splenic pulp pressure, abdominal aortic flow and portal systemic shunting. To measure splenic pulp pressure, a microtip pressure transducer was inserted into the spleen pulp. Abdominal aortic flow was measured by placing an ultrasonic Doppler flow probe around the abdominal aorta between the diaphragm and celiac artery. Portal systemic shunting was calculated by injection of fluorescent microspheres in to the splenic vein and determining the percentage accumulation of spheres in liver and pulmonary beds. Endothelin-1 hypo-response was evaluated by measuring the change in abdominal aortic flow in response to endothelin-1 intravenous administration. In addition, thoracic aorta endothelin-1 contraction was measured in 5 mm isolated thoracic aorta rings ex-vivo using an ADI small vessel myograph.
RESULTS: In wild type and iNOS-/- mice splenic pulp pressure increased from 7.5 ± 1.1 mmHg and 7.2 ± 1 mmHg to 25.4 ± 3.1 mmHg and 22 ± 4 mmHg respectively. In eNOS-/- mice splenic pulp pressure was increased after 1 d (P = NS), after which it decreased and by 7 d was not significantly elevated when compared to 7 d sham operated controls (6.9 ± 0.6 mmHg and 7.3 ± 0.8 mmHg respectively, P = 0.3). Abdominal aortic flow was increased by 80% and 73% in 7 d portal vein ligated wild type and iNOS when compared to shams, whereas there was no significant difference in 7 d portal vein ligated eNOS-/- mice when compared to shams. Endothelin-1 induced a rapid reduction in abdominal aortic blood flow in wild type, eNOS-/- and iNOS-/- sham mice (50% ± 8%, 73% ± 9% and 47% ± 9% respectively). Following portal vein ligation endothelin-1 reduction in blood flow was significantly diminished in each mouse group. Abdominal aortic flow was reduced by 19% ± 9%, 32% ± 10% and 9% ± 9% in wild type, eNOS-/- and iNOS-/- mice respectively.
CONCLUSION: Aberrant endothelin-1 response in murine portal hypertension is NOS isoform independent. Moreover, portal hypertension in the portal vein ligation model is independent of ET-1 function.
Collapse
MESH Headings
- Animals
- Aorta, Abdominal/drug effects
- Aorta, Abdominal/physiopathology
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/physiopathology
- Blood Flow Velocity
- Disease Models, Animal
- Endothelin-1/administration & dosage
- Hypertension, Portal/genetics
- Hypertension, Portal/metabolism
- Hypertension, Portal/physiopathology
- Injections, Intravenous
- Mice, Inbred C57BL
- Mice, Knockout
- Nitric Oxide/blood
- Nitric Oxide Synthase Type II/deficiency
- Nitric Oxide Synthase Type II/genetics
- Nitric Oxide Synthase Type III/deficiency
- Nitric Oxide Synthase Type III/genetics
- Portal Pressure/drug effects
- Regional Blood Flow
- Time Factors
- Vasoconstriction/drug effects
- Vasoconstrictor Agents/administration & dosage
Collapse
|
186
|
Ding R, Feng L, He L, Chen Y, Wen P, Fu Z, Lin C, Yang S, Deng X, Zeng J, Sun G. Peroxynitrite decomposition catalyst prevents matrix metalloproteinase-9 activation and neurovascular injury after hemoglobin injection into the caudate nucleus of rats. Neuroscience 2015; 297:182-93. [PMID: 25849612 DOI: 10.1016/j.neuroscience.2015.03.065] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Revised: 03/19/2015] [Accepted: 03/29/2015] [Indexed: 10/23/2022]
Abstract
Hemoglobin (Hb) is a major constituent of blood and a potent mediator of oxidative or nitrative stress after intracerebral hemorrhage (ICH). Our previous study demonstrated that Hb could induce abundant peroxynitrite (ONOO(-)) formation in vivo, which may be involved in the blood-brain barrier (BBB) disruption, however, the drug intervention is absent and also the underlying mechanism. Using an experimental stroke model by injecting Hb into the caudate nucleus of male Sprague-Dawley rats, we assessed the role of ONOO(-) decomposition catalyst, 5,10,15,20-tetrakis (4-sulfonatophenyl) porphyrinato iron(III) [FeTPPS] in the activation of MMP-9 and Hb-induced neurovascular injuries. 3-Nitrotyrosine (3-NT, as an index of ONOO(-) formation) and NF-κB expression was measured by western blot (WB) and immunohistochemistry (IHC)/immunofluorescence (IF). Activity of MMP was evaluated by in situ zymography. Neurovascular injury was assessed using zonula occludens-1 (ZO-1) by WB and IF, fibronectin (FN) and neuron-specific nuclear protein (NeuN) IHC. Perihematomal cell death was determined by TUNEL assay. Behavioral outcome was measured by modified neurological severity score (mNSS) test. At the injured striata, profuse 3-NT was produced and mainly expressed in neutrophils and microglia/macrophages. 3-NT formation significantly colocalized with nuclear factor-κB (NF-κB) expression. In situ zymography showed that gelatinase activity was mostly co-localized with neurons and blood vessel walls and partly with neutrophils and microglia/macrophages. Enhanced 3-NT production, NF-κB induction and MMP-9 activation were obviously reduced after FeTPPS treatment. Hb-induced injury to tight junction protein (ZO-1), basal lamina of FN-immunopositive microvasculature and neural cells was evidently ameliorated by FeTPPS. In addition, apoptotic cell numbers as well as behavioral deficits were also improved. The present study shows that the administration of the ONOO(-) decomposition catalyst FeTPPS protects against Hb-induced neurovascular injuries and improves neurological function, which possibly in part by suppressing MMP-9 activation.
Collapse
Affiliation(s)
- R Ding
- The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - L Feng
- The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - L He
- Department of Endocrinology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Y Chen
- The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China.
| | - P Wen
- The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Z Fu
- The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - C Lin
- The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - S Yang
- Department of Neurosurgery, Gaoqing Campus of Central Hospital of Zibo, Gaoqing People's Hospital, Gaoqing, Zibo 256300, Shandong, China
| | - X Deng
- Department of Neurosurgery, 999 Brain Hospital, Jinan University, Guangzhou 510510, China
| | - J Zeng
- The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - G Sun
- Department of Organ Transplantation, Zhujiang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
187
|
MicroRNA-532-3p regulates mitochondrial fission through targeting apoptosis repressor with caspase recruitment domain in doxorubicin cardiotoxicity. Cell Death Dis 2015; 6:e1677. [PMID: 25766316 PMCID: PMC4385919 DOI: 10.1038/cddis.2015.41] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 01/14/2015] [Accepted: 01/16/2015] [Indexed: 12/28/2022]
Abstract
Doxorubicin (DOX) is a wide-spectrum antitumor drug, but its clinical application is limited by its cardiotoxicity. However, the mechanisms underlying DOX-induced cardiomyopathy remain mostly unclear. Here we observed that apoptosis repressor with caspase recruitment domain (ARC) was downregulated in mouse heart and cardiomyocytes upon DOX treatment. Furthermore, enforced expression of ARC attenuated DOX-induced cardiomyocyte mitochondrial fission and apoptosis. ARC transgenic mice demonstrated reduced cardiotoxicity upon DOX administration. DOX-induced mitochondrial fission required the activity of dynamin-related protein 1 (Drp1). In elucidating the molecular mechanism by which ARC was downregulated upon DOX treatment, miR-532-3p was found to directly target ARC and participated in DOX-induced mitochondrial fission and apoptosis. MiR-532-3p was not involved in DOX-induced apoptosis in cancer cells. Taken together, these findings provide novel evidence that miR-532-3p and ARC constitute an antiapoptotic pathway that regulates DOX cardiotoxicity. Therefore, the development of new therapeutic strategies based on ARC and miR-532-3p is promising for overcoming the cardiotoxicity of chemotherapy for cancer therapy.
Collapse
|
188
|
Varga ZV, Giricz Z, Liaudet L, Haskó G, Ferdinandy P, Pacher P. Interplay of oxidative, nitrosative/nitrative stress, inflammation, cell death and autophagy in diabetic cardiomyopathy. BIOCHIMICA ET BIOPHYSICA ACTA 2015; 1852:232-242. [PMID: 24997452 PMCID: PMC4277896 DOI: 10.1016/j.bbadis.2014.06.030] [Citation(s) in RCA: 220] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Revised: 06/11/2014] [Accepted: 06/24/2014] [Indexed: 12/26/2022]
Abstract
Diabetes is a recognized risk factor for cardiovascular diseases and heart failure. Diabetic cardiovascular dysfunction also underscores the development of diabetic retinopathy, nephropathy and neuropathy. Despite the broad availability of antidiabetic therapy, glycemic control still remains a major challenge in the management of diabetic patients. Hyperglycemia triggers formation of advanced glycosylation end products (AGEs), activates protein kinase C, enhances polyol pathway, glucose autoxidation, which coupled with elevated levels of free fatty acids, and leptin have been implicated in increased generation of superoxide anion by mitochondria, NADPH oxidases and xanthine oxidoreductase in diabetic vasculature and myocardium. Superoxide anion interacts with nitric oxide forming the potent toxin peroxynitrite via diffusion limited reaction, which in concert with other oxidants triggers activation of stress kinases, endoplasmic reticulum stress, mitochondrial and poly(ADP-ribose) polymerase 1-dependent cell death, dysregulates autophagy/mitophagy, inactivates key proteins involved in myocardial calcium handling/contractility and antioxidant defense, activates matrix metalloproteinases and redox-dependent pro-inflammatory transcription factors (e.g. nuclear factor kappaB) promoting inflammation, AGEs formation, eventually culminating in myocardial dysfunction, remodeling and heart failure. Understanding the complex interplay of oxidative/nitrosative stress with pro-inflammatory, metabolic and cell death pathways is critical to devise novel targeted therapies for diabetic cardiomyopathy, which will be overviewed in this brief synopsis. This article is part of a Special Issue entitled: Autophagy and protein quality control in cardiometabolic diseases.
Collapse
Affiliation(s)
- Zoltán V Varga
- Laboratory of Physiological Studies, National Institutes of Health/NIAAA, Bethesda, MD, USA; Cardiometabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Zoltán Giricz
- Cardiometabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Lucas Liaudet
- Department of Intensive Care Medicine BH 08-621-University Hospital Medical Center 1011 LAUSANNE Switzerland
| | - György Haskó
- Department of Surgery and Center for Immunity and Inflammation, Rutgers NJ Medical School, USA
| | - Peter Ferdinandy
- Cardiometabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Pharmahungary Group, Szeged, Hungary
| | - Pál Pacher
- Laboratory of Physiological Studies, National Institutes of Health/NIAAA, Bethesda, MD, USA.
| |
Collapse
|
189
|
Meyer G, André L, Kleindienst A, Singh F, Tanguy S, Richard S, Obert P, Boucher F, Jover B, Cazorla O, Reboul C. Carbon monoxide increases inducible NOS expression that mediates CO-induced myocardial damage during ischemia-reperfusion. Am J Physiol Heart Circ Physiol 2015; 308:H759-67. [PMID: 25595132 DOI: 10.1152/ajpheart.00702.2014] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 01/13/2015] [Indexed: 11/22/2022]
Abstract
We investigated the role of inducible nitric oxide (NO) synthase (iNOS) on ischemic myocardial damage in rats exposed to daily low nontoxic levels of carbon monoxide (CO). CO is a ubiquitous environmental pollutant that impacts on mortality and morbidity from cardiovascular diseases. We have previously shown that CO exposure aggravates myocardial ischemia-reperfusion (I/R) injury partly because of increased oxidative stress. Nevertheless, cellular mechanisms underlying cardiac CO toxicity remain hypothetical. Wistar rats were exposed to simulated urban CO pollution for 4 wk. First, the effects of CO exposure on NO production and NO synthase (NOS) expression were evaluated. Myocardial I/R was performed on isolated perfused hearts in the presence or absence of S-methyl-isothiourea (1 μM), a NOS inhibitor highly specific for iNOS. Finally, Ca(2+) handling was evaluated in isolated myocytes before and after an anoxia-reoxygenation performed with or without S-methyl-isothiourea or N-acetylcystein (20 μM), a nonspecific antioxidant. Our main results revealed that 1) CO exposure altered the pattern of NOS expression, which is characterized by increased neuronal NOS and iNOS expression; 2) cardiac NO production increased in CO rats because of its overexpression of iNOS; and 3) the use of a specific inhibitor of iNOS reduced myocardial hypersensitivity to I/R (infarct size, 29 vs. 51% of risk zone) in CO rat hearts. These last results are explained by the deleterious effects of NO and reactive oxygen species overproduction by iNOS on diastolic Ca(2+) overload and myofilaments Ca(2+) sensitivity. In conclusion, this study highlights the involvement of iNOS overexpression in the pathogenesis of simulated urban CO air pollution exposure.
Collapse
Affiliation(s)
| | - Lucas André
- Institut national de la santé et de la recherche médicale, Université Montpellier1, Université Montpellier2, Montpellier, France
| | | | - François Singh
- Fédération de Médecine Translationelle, Faculty of Medicine, Université de Strasbourg, Strasbourg France
| | - Stéphane Tanguy
- Université d'Avignon, Avignon, France; Laboratoire Techniques for biomedical engineering and complexity management-informatics, mathematics, and applications-Grenoble, Bâtiment Jean Roget-Domaine de la Merci, Université Joseph Fourier, La Tronche Cedex, France
| | - Sylvain Richard
- Institut national de la santé et de la recherche médicale, Université Montpellier1, Université Montpellier2, Montpellier, France
| | | | - François Boucher
- Laboratoire Techniques for biomedical engineering and complexity management-informatics, mathematics, and applications-Grenoble, Bâtiment Jean Roget-Domaine de la Merci, Université Joseph Fourier, La Tronche Cedex, France
| | - Bernard Jover
- Centre de Pharmacologie et Innovation dans le Diabète, Faculty of Pharmacy, Université Montpellier1, Montpellier, France; and
| | - Olivier Cazorla
- Institut national de la santé et de la recherche médicale, Université Montpellier1, Université Montpellier2, Montpellier, France
| | | |
Collapse
|
190
|
Nair RR, Bagheri M, Saini DK. Temporally distinct roles of ATM and ROS in genotoxic-stress-dependent induction and maintenance of cellular senescence. J Cell Sci 2015; 128:342-53. [DOI: 10.1242/jcs.159517] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
ABSTRACT
Cells exposed to genotoxic stress induce cellular senescence through a DNA damage response (DDR) pathway regulated by ATM kinase and reactive oxygen species (ROS). Here, we show that the regulatory roles for ATM kinase and ROS differ during induction and maintenance of cellular senescence. Cells treated with different genotoxic agents were analyzed using specific pathway markers and inhibitors to determine that ATM kinase activation is directly proportional to the dose of the genotoxic stress and that senescence initiation is not dependent on ROS or the p53 status of cells. Cells in which ROS was quenched still activated ATM and initiated the DDR when insulted, and progressed normally to senescence. By contrast, maintenance of a viable senescent state required the presence of ROS as well as activated ATM. Inhibition or removal of either of the components caused cell death in senescent cells, through a deregulated ATM–ROS axis. Overall, our work demonstrates existence of an intricate temporal hierarchy between genotoxic stress, DDR and ROS in cellular senescence. Our model reports the existence of different stages of cellular senescence with distinct regulatory networks.
Collapse
Affiliation(s)
- Raji R. Nair
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore 560012, India
| | - Meisam Bagheri
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore 560012, India
| | - Deepak Kumar Saini
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore 560012, India
| |
Collapse
|
191
|
Hao E, Mukhopadhyay P, Cao Z, Erdélyi K, Holovac E, Liaudet L, Lee WS, Haskó G, Mechoulam R, Pacher P. Cannabidiol Protects against Doxorubicin-Induced Cardiomyopathy by Modulating Mitochondrial Function and Biogenesis. Mol Med 2015; 21:38-45. [PMID: 25569804 PMCID: PMC4461586 DOI: 10.2119/molmed.2014.00261] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 12/23/2014] [Indexed: 11/06/2022] Open
Abstract
Doxorubicin (DOX) is a widely used, potent chemotherapeutic agent; however, its clinical application is limited because of its dose-dependent cardiotoxicity. DOX's cardiotoxicity involves increased oxidative/nitrative stress, impaired mitochondrial function in cardiomyocytes/endothelial cells and cell death. Cannabidiol (CBD) is a nonpsychotropic constituent of marijuana, which is well tolerated in humans, with antioxidant, antiinflammatory and recently discovered antitumor properties. We aimed to explore the effects of CBD in a well-established mouse model of DOX-induced cardiomyopathy. DOX-induced cardiomyopathy was characterized by increased myocardial injury (elevated serum creatine kinase and lactate dehydrogenase levels), myocardial oxidative and nitrative stress (decreased total glutathione content and glutathione peroxidase 1 activity, increased lipid peroxidation, 3-nitrotyrosine formation and expression of inducible nitric oxide synthase mRNA), myocardial cell death (apoptotic and poly[ADP]-ribose polymerase 1 [PARP]-dependent) and cardiac dysfunction (decline in ejection fraction and left ventricular fractional shortening). DOX also impaired myocardial mitochondrial biogenesis (decreased mitochondrial copy number, mRNA expression of peroxisome proliferator-activated receptor γ coactivator 1-alpha, peroxisome proliferator-activated receptor alpha, estrogen-related receptor alpha), reduced mitochondrial function (attenuated complex I and II activities) and decreased myocardial expression of uncoupling protein 2 and 3 and medium-chain acyl-CoA dehydrogenase mRNA. Treatment with CBD markedly improved DOX-induced cardiac dysfunction, oxidative/nitrative stress and cell death. CBD also enhanced the DOX-induced impaired cardiac mitochondrial function and biogenesis. These data suggest that CBD may represent a novel cardioprotective strategy against DOX-induced cardiotoxicity, and the above-described effects on mitochondrial function and biogenesis may contribute to its beneficial properties described in numerous other models of tissue injury.
Collapse
Affiliation(s)
- Enkui Hao
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, United States of America
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Partha Mukhopadhyay
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Zongxian Cao
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Katalin Erdélyi
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Eileen Holovac
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Lucas Liaudet
- Department of Intensive Care Medicine, BH 08-621 University Hospital Medical Center, Lausanne, Switzerland
| | - Wen-Shin Lee
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, United States of America
- Division of General Medicine, Department of Medicine, Taipei Veterans General Hospital, National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - György Haskó
- Departments of Surgery, Rutgers New Jersey Medical School, Newark, New Jersey, United States of America
| | - Raphael Mechoulam
- Department for Medicinal Chemistry and Natural Products, Faculty of Medicine, Hebrew University of Jerusalem, Ein Kerem, Jerusalem, Israel
| | - Pál Pacher
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
192
|
Edwardson DW, Narendrula R, Chewchuk S, Mispel-Beyer K, Mapletoft JPJ, Parissenti AM. Role of Drug Metabolism in the Cytotoxicity and Clinical Efficacy of Anthracyclines. Curr Drug Metab 2015; 16:412-26. [PMID: 26321196 PMCID: PMC5398089 DOI: 10.2174/1389200216888150915112039] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 07/31/2015] [Accepted: 08/10/2015] [Indexed: 01/19/2023]
Abstract
Many clinical studies involving anti-tumor agents neglect to consider how these agents are metabolized within the host and whether the creation of specific metabolites alters drug therapeutic properties or toxic side effects. However, this is not the case for the anthracycline class of chemotherapy drugs. This review describes the various enzymes involved in the one electron (semi-quinone) or two electron (hydroxylation) reduction of anthracyclines, or in their reductive deglycosidation into deoxyaglycones. The effects of these reductions on drug antitumor efficacy and toxic side effects are also discussed. Current evidence suggests that the one electron reduction of anthracyclines augments both their tumor toxicity and their toxicity towards the host, in particular their cardiotoxicity. In contrast, the two electron reduction (hydroxylation) of anthracyclines strongly reduces their ability to kill tumor cells, while augmenting cardiotoxicity through their accumulation within cardiomyocytes and their direct effects on excitation/contraction coupling within the myocytes. The reductive deglycosidation of anthracyclines appears to inactivate the drug and only occurs under rare, anaerobic conditions. This knowledge has resulted in the identification of important new approaches to improve the therapeutic index of anthracyclines, in particular by inhibiting their cardiotoxicity. The true utility of these approaches in the management of cancer patients undergoing anthracycline-based chemotherapy remains unclear, although one such agent (the iron chelator dexrazoxane) has recently been approved for clinical use.
Collapse
Affiliation(s)
| | | | | | | | | | - Amadeo M Parissenti
- Dept. of Chemistry and Biochemistry, Laurentian University, 935 Ramsey Lake Road, Sudbury, ON P3E 2C6, Canada.
| |
Collapse
|
193
|
Gordiienko IА. Activity of trypsin-like enzymes and gelatinases in rats with doxorubicin cardiomyopathy. UKRAINIAN BIOCHEMICAL JOURNAL 2014. [DOI: 10.15407/ubj86.06.139] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
194
|
Nikitovic D, Juranek I, Wilks MF, Tzardi M, Tsatsakis A, Tzanakakis GN. Anthracycline-dependent cardiotoxicity and extracellular matrix remodeling. Chest 2014; 146:1123-1130. [PMID: 25288002 DOI: 10.1378/chest.14-0460] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
The mechanisms of anthracycline-dependent cardiotoxicity have been studied widely, with the suggested principal mechanism of anthracycline damage being the generation of reactive oxygen species by iron-anthracycline complexes, leading to lipid peroxidation and membrane damage. An increasing number of researchers studying cardiovascular events associated with anthracycline-based chemotherapy are addressing cardiac extracellular matrix (ECM) remodeling. The heart is an efficient muscular pump, with the cardiomyocytes and intramural coronary vasculature of the heart tethered in an ECM consisting of a network of fibrillar, structural proteins, mostly collagens. Increasing evidence suggests that the ECM plays a complex and diverse role in the processes initiated by anthracycline-class drugs that lead to cardiac damage. This review discusses adverse myocardial remodeling induced by anthracyclines and focuses on their mechanisms of action.
Collapse
Affiliation(s)
- Dragana Nikitovic
- Department of Anatomy-Histology-Embryology, School of Medicine, University of Crete, Heraklion, Greece.
| | - Ivo Juranek
- Institute of Experimental Pharmacology and Toxicology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Martin F Wilks
- Swiss Centre for Applied Human Toxicology, University of Basel, Basel, Switzerland
| | - Maria Tzardi
- Department of Pathology, School of Medicine, University of Crete, Heraklion, Greece
| | - Aristidis Tsatsakis
- Department of Forensic Sciences and Toxicology, School of Medicine, University of Crete, Heraklion, Greece
| | - George N Tzanakakis
- Department of Anatomy-Histology-Embryology, School of Medicine, University of Crete, Heraklion, Greece
| |
Collapse
|
195
|
Wang J, Hao L, Wang Y, Qin W, Wang X, Zhao T, Liu Y, Sheng L, Du Y, Zhang M, Lu Q. Inhibition of poly (ADP-ribose) polymerase and inducible nitric oxide synthase protects against ischemic myocardial damage by reduction of apoptosis. Mol Med Rep 2014; 11:1768-76. [PMID: 25412407 PMCID: PMC4270331 DOI: 10.3892/mmr.2014.2977] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2014] [Accepted: 09/19/2014] [Indexed: 01/12/2023] Open
Abstract
Myocardial infarction (MI) is defined as the deprivation of the myocardial tissue of oxygen and nutrients, resulting in the induction of inflammation and apoptosis of the cardiomyocytes. Poly (ADP-ribose) polymerase 1 (PARP1) is a nuclear enzyme closely associated with MI, that can be activated by DNA damage. Inducible nitric oxide synthase (iNOS) is a critical enzyme among the inflammatory cytokines. The present study aimed to investigate the underlying mechanism of the protective effects of PARP1 and iNOS inhibitor against MI, in rats. A total of 40 male Wistar rats were divided into four groups. The rats were anesthetized with sodium pentobarbital (50 mg/kg), and the left anterior descending coronary artery was occluded by ligation, using a 6-0 polypropylene monofilament suture, at the left atrial apex, in order to induce MI. The rats from each group received an abdominal injection of either dimethylsulfoxide (100 μl, for MI group); PARP-1 inhibitor, 3,4-dihydro-5-[4-(1-piperidinyl)butoxy]-1(2H)-isoquinolinone (DPQ; 10 mg/kg); or iNOS inhibitor, N-(1-naphthyl)ethylenediamine dihydrochloride (1400W; 10 mg/kg). The hearts were harvested from the rats after four weeks. Inhibition of PARP and iNOS activity improved heart function, as determined by serial echocardiography. The rate of apoptosis, as determined by a terminal deoxynucleotidyl-transferase-mediated dUTP nick end labeling assay, was reduced by 39.71 and 39.00% in the DPQ and 1400W groups, respectively, and this was accompanied by the downregulated expression of cleaved caspase-3 and PARP1. Effective inhibition of PARP and iNOS, by DPQ and 1400W, was detected by western blotting and immunofluorescence, and was shown to repress O2− and nitrotyrosine levels, following MI. The present study confirmed that inhibition of PARP1 and iNOS was able to protect against ischemic myocardial damage, by reducing the levels of apoptosis.
Collapse
Affiliation(s)
- Juan Wang
- Department of Cardiovascular Medicine, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Lin Hao
- Department of Cardiovascular Medicine, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Yan Wang
- Department of Anesthesiology, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250062, P.R. China
| | - Weidong Qin
- Department of Intensive Care Unit, Qilu Hospital Affiliated to Shandong University, Jinan, Shandong 250012, P.R. China
| | - Xin Wang
- Department of Cardiovascular Medicine, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Tong Zhao
- Department of Cardiovascular Medicine, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Yusheng Liu
- Department of Cardiovascular Medicine, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Lin Sheng
- Department of Cardiovascular Medicine, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Yimeng Du
- Department of Cardiovascular Medicine, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Mengyuan Zhang
- Department of Anesthesiology, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250062, P.R. China
| | - Qinghua Lu
- Department of Cardiovascular Medicine, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| |
Collapse
|
196
|
Dong Q, Chen L, Lu Q, Sharma S, Li L, Morimoto S, Wang G. Quercetin attenuates doxorubicin cardiotoxicity by modulating Bmi-1 expression. Br J Pharmacol 2014; 171:4440-54. [PMID: 24902966 PMCID: PMC4209150 DOI: 10.1111/bph.12795] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 04/29/2014] [Accepted: 05/15/2014] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND AND PURPOSE Doxorubicin-based chemotherapy induces cardiotoxicity, which limits its clinical application. We previously reported the protective effects of quercetin against doxorubicin-induced hepatotoxicity. In this study, we tested the effects of quercetin on the expression of Bmi-1, a protein regulating mitochondrial function and ROS generation, as a mechanism underlying quercetin-mediated protection against doxorubicin-induced cardiotoxicity. EXPERIMENTAL APPROACH Effects of quercetin on doxorubicin-induced cardiotoxicity was evaluated using H9c2 cardiomyocytes and C57BL/6 mice. Changes in apoptosis, mitochondrial function, oxidative stress and related signalling were evaluated in H9c2 cells. Cardiac function, serum enzyme activity and reactive oxygen species (ROS) generation were measured in mice after a single injection of doxorubicin with or without quercetin pre-treatment. KEY RESULTS In H9c2 cells, quercetin reduced doxorubicin-induced apoptosis, mitochondrial dysfunction, ROS generation and DNA double-strand breaks. The quercetin-mediated protection against doxorubicin toxicity was characterized by decreased expression of Bid, p53 and oxidase (p47 and Nox1) and by increased expression of Bcl-2 and Bmi-1. Bmi-1 siRNA abolished the protective effect of quercetin against doxorubicin-induced toxicity in H9c2 cells. Furthermore, quercetin protected mice from doxorubicin-induced cardiac dysfunction that was accompanied by reduced ROS levels and lipid peroxidation, but enhanced the expression of Bmi-1 and anti-oxidative superoxide dismutase. CONCLUSIONS AND IMPLICATIONS Our results demonstrate that quercetin decreased doxorubicin-induced cardiotoxicity in vitro and in vivo by reducing oxidative stress by up-regulation of Bmi-1 expression. The findings presented in this study have potential applications in preventing doxorubicin-induced cardiomyopathy.
Collapse
Affiliation(s)
- Qinghua Dong
- Central Lab of Biomedical Research Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang, China
| | - Long Chen
- Central Lab of Biomedical Research Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang, China
| | - Qunwei Lu
- Key Laboratory of Molecular Biophysics of Ministry of Education, School of Life Science and Technology, Huazhong University of Science and TechnologyWuhan, Hubei, China
| | - Sherven Sharma
- David Geffen School of Medicine at UCLA, and the Veterans AffairsLos Angeles, CA, USA
| | - Lei Li
- Department of Clinical Pharmacology, Graduate School of Medical Sciences, Kyushu UniversityFukuoka, Japan
| | - Sachio Morimoto
- Department of Clinical Pharmacology, Graduate School of Medical Sciences, Kyushu UniversityFukuoka, Japan
| | - Guanyu Wang
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang, China
| |
Collapse
|
197
|
Chen YL, Tsai YT, Lee CY, Lee CH, Chen CY, Liu CM, Chen JJ, Loh SH, Tsai CS. Urotensin II inhibits doxorubicin-induced human umbilical vein endothelial cell death by modulating ATF expression and via the ERK and Akt pathway. PLoS One 2014; 9:e106812. [PMID: 25268131 PMCID: PMC4182104 DOI: 10.1371/journal.pone.0106812] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 07/14/2014] [Indexed: 12/30/2022] Open
Abstract
Background and Purpose Regulation of the homeostasis of vascular endothelium is critical for the processes of vascular remodeling and angiogenesis under physiological and pathological conditions. Urotensin II (U-II), a potent vasoactive peptide, participates in vascular and myocardial remodeling after injury. We investigated the protective effect of U-II on doxorubicin (DOX)-induced apoptosis in cultured human umbilical vein endothelial cells (HUVECs) and the potential mechanisms involved in this process. Experimental Approach Cultured HUVECs were treated with vehicle, DOX (1 µM), U-II, or U-II plus DOX. Apoptosis was evaluated by DNA strand break level with TdT-mediated dUTP nick-end labeling (TUNEL) staining. Western blot analysis was employed to determine the related protein expression and flow cytometry assay was used to determine the TUNEL positive cells. Key Results U-II reduced the quantity of cleaved caspase-3 and cytosol cytochrome c and increased Bcl-2 expression, which results in protecting HUVECs from DOX-induced apoptosis. U-II induced Activating transcription factor 3 (ATF3) at both mRNA and protein levels in U-II-treated cells. Knockdown of ATF3 with ATF3 siRNA significantly reduced ATF3 protein levels and U-II protective effect under DOX-treated condition. U-II downregulated p53 expression in DOX-induced HUVECs apoptosis, and it rapidly activated extracellular signal-regulated protein kinase (ERK) and Akt. The DOX induced change of p53 was not affected by U-II antagonist (urantide) under ATF-3 knockdown. The inhibitory effect of U-II on DOX-increased apoptosis was attenuated by inhibitors of ERK (U0126) and PI3K/Akt (LY294002). Conclusion and Implications Our observations provide evidence that U-II protects HUVECs from DOX-induced apoptosis. ERK-Akt phosphorylation, ATF3 activation, and p53 downregulation may play a signal-transduction role in this process.
Collapse
Affiliation(s)
- Yen-Ling Chen
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Yi-Ting Tsai
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, Republic of China
- Division of Cardiovascular Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Chung-Yi Lee
- Division of Cardiovascular Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Chien-Hsing Lee
- Department of Nursing, Min-Hwei College of Health Care Management, Tainan, Taiwan, Republic of China
| | - Chung-Yi Chen
- School of Medical and Health Sciences, Fooyin University, Kaohsiung, Taiwan, Republic of China
| | - Chi-Ming Liu
- School of Medical and Health Sciences, Fooyin University, Kaohsiung, Taiwan, Republic of China
| | - Jin-Jer Chen
- Division of Cardiology, Department of Internal Medicine and Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan, Republic of China
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan, Republic of China
- Division of Cardiology, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan, Republic of China
| | - Shih-Hurng Loh
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, Republic of China
- Department of Pharmacology, National Defense Medical Center, Taipei, Taiwan, Republic of China
- * E-mail: (C-ST); (S-HL)
| | - Chien-Sung Tsai
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, Republic of China
- Division of Cardiovascular Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, Republic of China
- * E-mail: (C-ST); (S-HL)
| |
Collapse
|
198
|
Croteau E, Tremblay S, Gascon S, Dumulon-Perreault V, Labbé SM, Rousseau JA, Cunnane SC, Carpentier AC, Bénard F, Lecomte R. [(11)C]-Acetoacetate PET imaging: a potential early marker for cardiac heart failure. Nucl Med Biol 2014; 41:863-70. [PMID: 25195015 DOI: 10.1016/j.nucmedbio.2014.08.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2014] [Accepted: 08/04/2014] [Indexed: 10/24/2022]
Abstract
UNLABELLED The ketone body acetoacetate could be used as an alternate nutrient for the heart, and it also has the potential to improve cardiac function in an ischemic-reperfusion model or reduce the mitochondrial production of oxidative stress involved in cardiotoxicity. In this study, [(11)C]-acetoacetate was investigated as an early marker of intracellular damage in heart failure. METHODS A rat cardiotoxicity heart failure model was induced by doxorubicin, Dox(+). [(14)C]-Acetoacetate, a non-positron (β-) emitting radiotracer, was used to characterize the arterial blood input function and myocardial mitochondrial uptake. Afterward, [(11)C]-acetoacetate (β+) myocardial PET images were obtained for kinetic analysis and heart function assessment in control Dox(-) (n=15) and treated Dox(+) (n=6) rats. The uptake rate (K1) and myocardial clearance rate (k2or kmono) were extracted. RESULTS [(14)C]-Acetoacetate in the blood was increased in Dox(+), from 2 min post-injection until the last withdrawal point when the heart was harvested, as well as the uptake in the heart and myocardial mitochondria (unpaired t-test, p <0.05). PET kinetic analysis of [(11)C]-acetoacetate showed that rate constants K1, k2 and kmono were decreased in Dox(+) (p <0.05) combined with a reduction of 24% of the left ventricular ejection fraction (p <0.001). CONCLUSION Radioactive acetoacetate ex vivo analysis [(14)C], and in vivo kinetic [(11)C] studies provided evidence that [(11)C]-acetoacetate can assess heart failure Dox(+). Contrary to myocardial flow reserve (rest-stress protocol), [(11)C]-acetoacetate can be used to assess reduced kinetic rate constants without requirement of hyperemic stress response. The proposed [(11)C]-acetoacetate cardiac radiotracer in the investigation of heart disease is novel and paves the way to a potential role for [(11)C]-acetoacetate in cardiac pathophysiology.
Collapse
Affiliation(s)
- Etienne Croteau
- Sherbrooke Molecular Imaging Center of CRCHUS, Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, Sherbrooke, QC, Canada.
| | - Sébastien Tremblay
- Sherbrooke Molecular Imaging Center of CRCHUS, Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Suzanne Gascon
- Sherbrooke Molecular Imaging Center of CRCHUS, Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Véronique Dumulon-Perreault
- Sherbrooke Molecular Imaging Center of CRCHUS, Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Sébastien M Labbé
- Department of Medicine, Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - Jacques A Rousseau
- Sherbrooke Molecular Imaging Center of CRCHUS, Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Stephen C Cunnane
- Research Center on Aging, Université de Sherbrooke, Sherbrooke, QC, Canada; Department of Medicine, Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - André C Carpentier
- Department of Medicine, Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - François Bénard
- Division of Nuclear Medicine, Department of Radiology, University of British Columbia, Vancouver, BC, Canada
| | - Roger Lecomte
- Sherbrooke Molecular Imaging Center of CRCHUS, Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
199
|
Khan MA, Singh M, Khan MS, Najmi AK, Ahmad S. Caspase mediated synergistic effect of Boswellia serrata extract in combination with doxorubicin against human hepatocellular carcinoma. BIOMED RESEARCH INTERNATIONAL 2014; 2014:294143. [PMID: 25177685 PMCID: PMC4142179 DOI: 10.1155/2014/294143] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 06/20/2014] [Accepted: 06/23/2014] [Indexed: 02/02/2023]
Abstract
The study investigated the growth-inhibiting and apoptosis mediating effects of B. serrata extract as monotherapy and combination therapy with DOX against hepatocellular carcinoma cell lines. Boswellic acid rich fraction of B. serrata extract was prepared. MTT assay on HepG2 and Hep3B cells was carried out using B. serrata alone and in combination with DOX. Further, caspase-3 activity, TNF-α level, and IL-6 level were estimated. Isobolographic analysis was carried out to evaluate the effect of combination therapy. Additionally, protective effect of B. serrata extract on DOX induced hepatic toxicity was also evaluated in Wistar rats. B. serrata extract inhibited growth of HepG2 (IC50 value of 21.21 ± 0.92 μg/mL) as well as HepG2 (IC50 value of 18.65 ± 0.71 μg/mL). DOX inhibited growth in HepG2 and Hep3B cells with an IC50 of 1.06 ± 0.04 μg/mL and 1.92 ± 0.09 μg/mL. Isobolographic analysis showed combination index (CI) of DOX and B. serrata extract of 0.53 ± 0.03 to 0.79 ± 0.02 suggesting synergistic behavior against the two cell lines. B. serrata extract also caused dose dependent increase in caspase-3 activity, TNF-α level, and IL-6 level which was higher (P < 0.001) with DOX (1 μM) and B. serrata extract (20 μg/mL) combination. B. serrata extract also protected Wistar rats against DOX induced hepatic toxicity.
Collapse
Affiliation(s)
- Mohammad Ahmed Khan
- Department of Pharmacology, Faculty of Pharmacy, Hamdard University, Hamdard Nagar, New Delhi 110062, India
| | - Mhaveer Singh
- Department of Pharmacognosy & Phytochemistry, Faculty of Pharmacy, Hamdard University (Jamia Hamdard), Hamdard Nagar, New Delhi 110062, India
| | - Masood Shah Khan
- Department of Pharmacognosy & Phytochemistry, Faculty of Pharmacy, Hamdard University (Jamia Hamdard), Hamdard Nagar, New Delhi 110062, India
| | - Abul Kalam Najmi
- Department of Pharmacology, Faculty of Pharmacy, Hamdard University, Hamdard Nagar, New Delhi 110062, India
| | - Sayeed Ahmad
- Department of Pharmacognosy & Phytochemistry, Faculty of Pharmacy, Hamdard University (Jamia Hamdard), Hamdard Nagar, New Delhi 110062, India
| |
Collapse
|
200
|
Zhou S, Wang Y, Tan Y, Cai X, Cai L, Cai J, Zheng Y. Deletion of metallothionein exacerbates intermittent hypoxia-induced oxidative and inflammatory injury in aorta. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014; 2014:141053. [PMID: 25177426 PMCID: PMC4142187 DOI: 10.1155/2014/141053] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 07/08/2014] [Accepted: 07/08/2014] [Indexed: 01/18/2023]
Abstract
The present study was to explore the effect of metallothionein (MT) on intermittent hypoxia (IH) induced aortic pathogenic changes. Markers of oxidative damages, inflammation, and vascular remodeling were observed by immunohistochemical staining after 3 days and 1, 3, and 8 weeks after IH exposures. Endogenous MT was induced after 3 days of IH but was significantly decreased after 8 weeks of IH. Compared with the wild-type mice, MT knock-out mice exhibited earlier and more severe pathogenic changes of oxidative damages, inflammatory responses, and cellular apoptosis, as indicated by the significant accumulation of collagen, increased levels of connective tissue growth factor, transforming growth factor β1, tumor necrosis factor-alpha, vascular cell adhesion molecule 1,3-nitrotyrosine, and 4-hydroxy-2-nonenal in the aorta. These findings suggested that chronic IH may lead to aortic damages characterized by oxidative stress and inflammation, and MT may play a pivotal role in the above pathogenesis process.
Collapse
Affiliation(s)
- Shanshan Zhou
- The Center of Cardiovascular Diseases at the First Hospital of Jilin University, 71 Xinmin Street, Changchun 130021, China
- Kosair Children's Hospital Research Institute at the Department of Pediatrics, University of Louisville, 570 South Preston Street, Baxter I, Suite 321B, Louisville, KY 40202, USA
| | - Yonggang Wang
- The Center of Cardiovascular Diseases at the First Hospital of Jilin University, 71 Xinmin Street, Changchun 130021, China
- Kosair Children's Hospital Research Institute at the Department of Pediatrics, University of Louisville, 570 South Preston Street, Baxter I, Suite 321B, Louisville, KY 40202, USA
| | - Yi Tan
- Kosair Children's Hospital Research Institute at the Department of Pediatrics, University of Louisville, 570 South Preston Street, Baxter I, Suite 321B, Louisville, KY 40202, USA
- Chinese-American Research Institute for Diabetic Complication, Wenzhou Medical College, Wenzhou 325035, China
| | - Xiaohong Cai
- Department of Pediatrics, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Lu Cai
- Kosair Children's Hospital Research Institute at the Department of Pediatrics, University of Louisville, 570 South Preston Street, Baxter I, Suite 321B, Louisville, KY 40202, USA
- Departments of Radiation Oncology and Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA
| | - Jun Cai
- Kosair Children's Hospital Research Institute at the Department of Pediatrics, University of Louisville, 570 South Preston Street, Baxter I, Suite 321B, Louisville, KY 40202, USA
| | - Yang Zheng
- The Center of Cardiovascular Diseases at the First Hospital of Jilin University, 71 Xinmin Street, Changchun 130021, China
| |
Collapse
|