201
|
Boghdady NAE. Antioxidant and antiapoptotic effects of proanthocyanidin and ginkgo biloba extract against doxorubicin-induced cardiac injury in rats. Cell Biochem Funct 2012; 31:344-51. [DOI: 10.1002/cbf.2907] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Revised: 07/22/2012] [Accepted: 09/10/2012] [Indexed: 12/21/2022]
|
202
|
Carbone A, Psaltis PJ, Nelson AJ, Metcalf R, Richardson JD, Weightman M, Thomas A, Finnie JW, Young GD, Worthley SG. Dietary omega-3 supplementation exacerbates left ventricular dysfunction in an ovine model of anthracycline-induced cardiotoxicity. J Card Fail 2012; 18:502-11. [PMID: 22633309 DOI: 10.1016/j.cardfail.2012.03.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2012] [Revised: 03/26/2012] [Accepted: 03/28/2012] [Indexed: 10/28/2022]
Abstract
BACKGROUND Cumulative dose-dependent nonischemic cardiomyopathy (NICM) remains a significant risk with the use of some chemotherapeutic agents. In this context, omega-3 polyunsaturated fatty acids (PUFA) have been investigated for their cardioprotective potential in rodent and in vitro models of anthracycline toxicity, with conflicting results. This study evaluated prophylactic omega-3 PUFA supplementation in a large-animal model of anthracycline-induced NICM. METHODS AND RESULTS Merino sheep were randomized to oral drenching with omega-3 PUFA (fish oil; n = 8) or olive oil placebo (n = 9) 3 weeks before commencing repeated intracoronary infusions of doxorubicin (DOX) to induce cardiac dysfunction. Cumulative DOX dose was 3.6 mg/kg. Drenching was continued for 12 weeks after final DOX exposure. Despite significant increases in tissue omega-3 PUFA levels (P < .05 vs placebo), omega-3-treated sheep displayed greater signs of anthracycline cardiotoxicity than placebo animals, consisting of left ventricular dilatation and a greater decline in ejection fraction (P < .05), although myocardial fibrosis burden was similar in both groups. CONCLUSIONS Dietary intake of omega-3 PUFA fails to prevent and may indeed exacerbate DOX-induced cardiotoxicity. Clinical use of omega-3 supplementation during chemotherapy should be deferred until more information is available regarding the mechanisms of interaction between fatty acids and the myocardium during anthracycline exposure.
Collapse
Affiliation(s)
- Angelo Carbone
- Discipline of Medicine, Faculty of Health Sciences, University of Adelaide and Cardiovascular Research Centre, Royal Adelaide Hospital, Adelaide, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
203
|
Chorvatova A, Elzwiei F, Mateasik A, Chorvat D. Effect of ouabain on metabolic oxidative state in living cardiomyocytes evaluated by time-resolved spectroscopy of endogenous NAD(P)H fluorescence. JOURNAL OF BIOMEDICAL OPTICS 2012; 17:101505. [PMID: 23223981 DOI: 10.1117/1.jbo.17.10.101505] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Time-resolved spectrometry of endogenous nicotinamide dinucleotide phosphate [NAD(P)H] fluorescence is a useful method to evaluate metabolic oxidative state in living cells. Ouabain is a well-known pharmaceutical drug used in the treatment of cardiovascular disease, the effects of which on myocardial metabolism were recently demonstrated. Mechanisms implicated in these actions are still poorly understood. We investigate the effect of ouabain on the metabolic oxidative state of living cardiac cells identified by time-resolved fluorescence spectroscopy of mitochondrial NAD(P)H. Spectral unmixing is used to resolve individual NAD(P)H fluorescence components. Ouabain decreased the integral intensity of NAD(P)H fluorescence, leading to a reduced component amplitudes ratio corresponding to a change in metabolic state. We also noted that lactate/pyruvate, affecting the cytosolic NADH gradient, increased the effect of ouabain on the component amplitudes ratio. Cell oxidation levels, evaluated as the percentage of oxidized NAD(P)H, decreased exponentially with rising concentrations of the cardiac glycoside. Ouabain also stimulated the mitochondrial NADH production. Our study sheds a new light on the role that ouabain plays in the regulation of metabolic state, and presents perspective on a noninvasive, pharmaceutical approach for testing the effect of drugs on the mitochondrial metabolism by means of time-resolved fluorescence spectroscopy in living cells.
Collapse
Affiliation(s)
- Alzbeta Chorvatova
- Department of Biophotonics, International Laser Center, 84104 Bratislava, Slovakia.
| | | | | | | |
Collapse
|
204
|
Intensification of doxorubicin-related oxidative stress in the heart by hypothyroidism is not related to the expression of cytochrome P450 NADPH-reductase and inducible nitric oxide synthase, as well as activity of xanthine oxidase. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2012; 2012:139327. [PMID: 22966413 PMCID: PMC3432562 DOI: 10.1155/2012/139327] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Accepted: 07/05/2012] [Indexed: 11/17/2022]
Abstract
Cytochrome P450 NADPH-reductase (P450R), inducible synthase (iNOS) and xanthine oxidase play an important role in the antracycline-related cardiotoxicity. The expression of P450R and iNOS is regulated by triiodothyronine. The aim of this study was to evaluate the effect of methimazole-induced hypothyreosis on oxidative stress secondary to doxorubicin administration. 48 hours after methimazole giving cessation, rats were exposed to doxorubicin (2.0, 5.0 and 15 mg/kg). Blood and heart were collected 4, 48 and 96 h after the drug administration. Animals exposed exclusively to doxorubicin or untreated ones were also assessed. The hypothyreosis (0.025% of methimazole) significantly increased the doxorubicin effect on the cardiac carbonyl group and they may increase the glutathione level. An insignificant effect of methimazole was noticed in case of the cardiac lipid peroxidation product, the amount of DNA oxidative damages, iNOS and xanthine oxidase-enzymes responsible for red-ox activation of doxorubicin. However, the concentration of P450R was affected by a lower dose of methimazole in rats administered with doxorubicin. Since in rats receiving doxorubicin changes in oxidative stress caused by methimazole were not accompanied by elevation of bioreductive enzymes, it may be concluded that these changes in the oxidative stress were not related to the tested enzymes.
Collapse
|
205
|
Jirkovsky E, Popelová O, Kriváková-Stanková P, Vávrová A, Hroch M, Hasková P, Brcáková-Dolezelová E, Micuda S, Adamcová M, Simůnek T, Cervinková Z, Gersl V, Sterba M. Chronic Anthracycline Cardiotoxicity: Molecular and Functional Analysis with Focus on Nuclear Factor Erythroid 2-Related Factor 2 and Mitochondrial Biogenesis Pathways. J Pharmacol Exp Ther 2012; 343:468-78. [PMID: 22915767 DOI: 10.1124/jpet.112.198358] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Affiliation(s)
- Eduard Jirkovsky
- Department of Pharmacology, Faculty of Medicine in Hradec Králové, Charles University in Prague, Šimkova 870, Hradec Králové, 500 38, Czech Republic
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
206
|
Drug-induced oxidative stress and toxicity. J Toxicol 2012; 2012:645460. [PMID: 22919381 PMCID: PMC3420138 DOI: 10.1155/2012/645460] [Citation(s) in RCA: 398] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Revised: 04/26/2012] [Accepted: 04/29/2012] [Indexed: 12/14/2022] Open
Abstract
Reactive oxygen species (ROS) are a byproduct of normal metabolism and have roles in cell signaling and homeostasis. Species include oxygen radicals and reactive nonradicals. Mechanisms exist that regulate cellular levels of ROS, as their reactive nature may otherwise cause damage to key cellular components including DNA, protein, and lipid. When the cellular antioxidant capacity is exceeded, oxidative stress can result. Pleiotropic deleterious effects of oxidative stress are observed in numerous disease states and are also implicated in a variety of drug-induced toxicities. In this paper, we examine the nature of ROS-induced damage on key cellular targets of oxidative stress. We also review evidence implicating ROS in clinically relevant, drug-related side effects including doxorubicin-induced cardiac damage, azidothymidine-induced myopathy, and cisplatin-induced ototoxicity.
Collapse
|
207
|
Nawara K, Krysinski P, Blanchard GJ. Photoinduced Reactivity of Doxorubicin: Catalysis and Degradation. J Phys Chem A 2012; 116:4330-7. [DOI: 10.1021/jp303218r] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Krzysztof Nawara
- Department of Chemistry, University of Warsaw, Pasteura 1, Warsaw 02-093 Poland
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824,
United States
| | - Pawel Krysinski
- Department of Chemistry, University of Warsaw, Pasteura 1, Warsaw 02-093 Poland
| | - G. J. Blanchard
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824,
United States
| |
Collapse
|
208
|
Malhi SS, Budhiraja A, Arora S, Chaudhari KR, Nepali K, Kumar R, Sohi H, Murthy RSR. Intracellular delivery of redox cycler-doxorubicin to the mitochondria of cancer cell by folate receptor targeted mitocancerotropic liposomes. Int J Pharm 2012; 432:63-74. [PMID: 22531856 DOI: 10.1016/j.ijpharm.2012.04.030] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Revised: 04/04/2012] [Accepted: 04/08/2012] [Indexed: 11/29/2022]
Abstract
Cancer cells reflect higher level of ROS in comparison to the normal cell, so they become more vulnerable to further oxidative stress induced by exogenous ROS-generating agents. Through this a novel therapeutic strategy has evolved, which involves the delivery of redox cycler-doxorubicin (DOX) to the mitochondria of cancer cell where it acts as a source of exogenous ROS production. The purpose of this study is to develop a liposomal preparation which exhibits a propensity to selectively target cancer cell along with the potential of delivering drug to mitochondria of cell. We have rendered liposomes mitocancerotropic (FA-MTLs) by their surface modification with dual ligands, folic acid (FA) for cancer cell targeting and triphenylphosphonium (TPP) cations for mitochondria targeting. The cytotoxicity, ROS production and cell uptake of doxorubicin loaded liposomes were evaluated in FR (+) KB cells and found to be increased considerably with FA-MTLs in comparison to folic acid appended, mitochondria targeted and non-targeted liposomes. As confirmed by confocal microscopy, the STPP appended liposomes delivered DOX to mitochondria of cancer cell and also showed higher ROS production and cytotoxicity in comparison to folic acid appended and non-targeted liposomes. Most importantly, mitocancerotropic liposomes showed superior activity over mitochondria targeted liposomes which confirm the synergistic effect imparted by the presence of dual ligands - folic acid and TPP on the enhancement of cellular and mitochondrial delivery of doxorubicin in KB cells.
Collapse
Affiliation(s)
- Sarandeep Singh Malhi
- Nano-Medicine Research Center, Department of Pharmaceutics, Indo-Soviet Friendship College of Pharmacy, Moga, Punjab, India
| | | | | | | | | | | | | | | |
Collapse
|
209
|
Dietary inorganic nitrate alleviates doxorubicin cardiotoxicity: mechanisms and implications. Nitric Oxide 2012; 26:274-84. [PMID: 22484629 DOI: 10.1016/j.niox.2012.03.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Revised: 02/11/2012] [Accepted: 03/22/2012] [Indexed: 11/21/2022]
Abstract
Doxorubicin (DOX) is one of the most powerful and widely prescribed chemotherapeutic agents to treat divergent human cancers. However, the clinical use of DOX is restricted due to its severe cardiotoxic side-effects. There has been ongoing search for cardioprotectants against DOX toxicity. Inorganic nitrate has emerged as a bioactive compound that can be reduced into nitrite and nitric oxide in vivo and in turn plays a therapeutic role in diseases associated with nitric oxide insufficiency or dysregulation. In this review, we describe a novel concept of using dietary supplementation of inorganic nitrate to reduce DOX-induced cardiac cellular damage and dysfunction, based on our recent promising studies in a mouse model of DOX cardiotoxicity. Our data show that chronic oral ingestion of sodium nitrate, at a dose equivalent to ~400% of the Acceptable Daily Intake of the World Health Organization, alleviated DOX-induced left ventricular dysfunction and mitochondrial respiratory chain damage. Such cardioprotective effects were associated with reduction of cardiomyocyte necrosis/apoptosis, tissue lipid peroxidation, and mitochondrial H(2)O(2) generation following DOX treatment. Furthermore, proteomic studies revealed enhanced cardiac expression of mitochondrial antioxidant enzyme - peroxiredoxin 5 in the nitrate-treated animals. These studies suggest that inorganic nitrate could be an inexpensive therapeutic agent for long-term oral administration in preventing DOX-induced cardiac toxicity and myopathy during the prolonged pathological process. Future clinical trials in the cancer patients undergoing DOX chemotherapy are warranted to translate these experimental findings into an effective new therapy in preventing the DOX-induced cardiomyopathy.
Collapse
|
210
|
Savatier J, Rharass T, Canal C, Gbankoto A, Vigo J, Salmon JM, Ribou AC. Adriamycin dose and time effects on cell cycle, cell death, and reactive oxygen species generation in leukaemia cells. Leuk Res 2012; 36:791-8. [PMID: 22417651 DOI: 10.1016/j.leukres.2012.02.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Revised: 01/10/2012] [Accepted: 02/20/2012] [Indexed: 01/03/2023]
Abstract
We investigate the relative importance of the different mechanisms of Adriamycin, an anthracycline, and their interrelations, in particular the link between cell cycle arrest, cell death, and generation of reactive oxygen species (ROS) that is suspected to be the origin of cardiotoxic side-effects. We introduced a lifetime fluorescence based technology and used videomicrofluorometry, two efficient analytical methods. We show that depending on the doses and time after incubation, ADR will not reach the same compartments (nucleus, mitochondria, cytosol) in the cells, having consequences on the production of ROS, growth arrest pathways and cell death pathways.
Collapse
Affiliation(s)
- Julien Savatier
- Institut Fresnel, Domaine Universitaire de St Jerôme, Marseille, France
| | | | | | | | | | | | | |
Collapse
|
211
|
The redox imbalance and the reduction of contractile protein content in rat hearts administered with L-thyroxine and Doxorubicin. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2012; 2012:681367. [PMID: 22530076 PMCID: PMC3317061 DOI: 10.1155/2012/681367] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2011] [Revised: 10/15/2011] [Accepted: 11/15/2011] [Indexed: 01/12/2023]
Abstract
Oxidative stress and disorders in calcium balance play a crucial role in the doxorubicin-induced cardiotoxicity. Moreover, many cardiotoxic targets of doxorubicin are regulated by iodothyronine hormones. The aim of the study was to evaluate effects of tetraiodothyronine (0.2, 2 mg/L) on oxidative stress in the cardiac muscle as well as contractility and cardiomyocyte damage markers in rats receiving doxorubicin (1.5 mg/kg) once a week for ten weeks. Doxorubicin was administered alone (DOX) or together with a lower (0.2T4 + DOX) and higher dose of tetraiodothyronine (2T4 + DOX). Two groups received only tetraiodothyronine (0.2T4, 2T4). Coadministration of tetraiodothyronine and doxorubicin increased the level of lipid peroxidation products and reduced RyR2 level when compared to untreated control and group exposed exclusively to doxorubicin. Insignificant differences in SERCA2 and occasional histological changes were observed. In conclusion, an increase of tetraiodothyronine level may be an additional risk factor of redox imbalance and RyR2 reduction in anthracycline cardiotoxicity.
Collapse
|
212
|
Salvatorelli E, Menna P, Gonzalez Paz O, Surapaneni S, Aukerman SL, Chello M, Covino E, Sung V, Minotti G. Pharmacokinetic characterization of amrubicin cardiac safety in an ex vivo human myocardial strip model. II. Amrubicin shows metabolic advantages over doxorubicin and epirubicin. J Pharmacol Exp Ther 2012; 341:474-83. [PMID: 22338034 DOI: 10.1124/jpet.111.190264] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Anthracycline-related cardiotoxicity correlates with cardiac anthracycline accumulation and bioactivation to secondary alcohol metabolites or reactive oxygen species (ROS), such as superoxide anion (O₂·⁻) and hydrogen peroxide H₂O₂). We reported that in an ex vivo human myocardial strip model, 3 or 10 μM amrubicin [(7S,9S)-9-acetyl-9-amino-7-[(2-deoxy-β-D-erythro-pentopyranosyl)oxy]-7,8,9,10-tetrahydro-6,11-dihydroxy-5,12-napthacenedione hydrochloride] accumulated to a lower level compared with equimolar doxorubicin or epirubicin (J Pharmacol Exp Ther 341:464-473, 2012). We have characterized how amrubicin converted to ROS or secondary alcohol metabolite in comparison with doxorubicin (that formed both toxic species) or epirubicin (that lacked ROS formation and showed an impaired conversion to alcohol metabolite). Amrubicin and doxorubicin partitioned to mitochondria and caused similar elevations of H₂O₂, but the mechanisms of H₂O₂ formation were different. Amrubicin produced H₂O₂ by enzymatic reduction-oxidation of its quinone moiety, whereas doxorubicin acted by inducing mitochondrial uncoupling. Moreover, mitochondrial aconitase assays showed that 3 μM amrubicin caused an O₂·⁻-dependent reversible inactivation, whereas doxorubicin always caused an irreversible inactivation. Low concentrations of amrubicin therefore proved similar to epirubicin in sparing mitochondrial aconitase from irreversible inactivation. The soluble fraction of human myocardial strips converted doxorubicin and epirubicin to secondary alcohol metabolites that irreversibly inactivated cytoplasmic aconitase; in contrast, strips exposed to amrubicin failed to generate its secondary alcohol metabolite, amrubicinol, and only occasionally exhibited an irreversible inactivation of cytoplasmic aconitase. This was caused by competing pathways that favored formation and complete or near-to-complete elimination of 9-deaminoamrubicinol. These results characterize amrubicin metabolic advantages over doxorubicin and epirubicin, which may correlate with amrubicin cardiac safety in preclinical or clinical settings.
Collapse
|
213
|
Carbonic anhydrase IX is a predictive marker of doxorubicin resistance in early-stage breast cancer independent of HER2 and TOP2A amplification. Br J Cancer 2012; 106:916-22. [PMID: 22333602 PMCID: PMC3305967 DOI: 10.1038/bjc.2012.32] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background: In early-stage breast cancer, adjuvant chemotherapy is associated with significant systemic toxicity with only a modest survival benefit. Therefore, there is considerable interest in identifying predictive markers of response to therapy. Doxorubicin, one of the most common drugs used to treat breast cancer, is an anthracycline chemotherapeutic agent, a class of drugs known to be affected by hypoxia. Accordingly, we examined whether expression of the endogenous hypoxia marker carbonic anhydrase IX (CA IX) is predictive of outcome in early-stage breast cancer patients treated with doxorubicin. Methods: We obtained 209 early-stage pre-treatment surgically-resected breast tumours from patients, who received doxorubicin in their chemotherapeutic regimen and had >10 years of follow-up. Immunohistochemistry was used to detect CA IX, and we used fluorescence in situ hybridisation to detect both human epidermal growth factor receptor (HER2) and DNA topoisomerase II-alpha (TOP2A) gene amplification. Results: Carbonic anhydrase IX intensity was significantly correlated with progression-free survival (PFS) and overall survival (OS) in patients receiving 300 mg m−2 of doxorubicin (HR=1.82 and 3.77; P=0.0014 and 0.010, respectively). There was a significant, inverse correlation between CA IX score and oestrogen receptor expression, but no significant correlations were seen with either HER2 or TOP2A ratio. Conclusion: We demonstrate that CA IX expression is correlated with worse PFS and OS for breast cancer patients treated with doxorubicin, independent of HER2 or TOP2A gene amplification. This study provides evidence that using CA IX to detect hypoxia in surgically-resected breast tumours may be of clinical use in choosing an appropriate chemotherapy regimen.
Collapse
|
214
|
Hanušová V, Boušová I, Pakostová A, Skálová L. The influence of oracin on reduction and toxicity of doxorubicin in hepatocytes and mammary epithelial cells MCF-10A. Xenobiotica 2012; 42:571-9. [PMID: 22217270 DOI: 10.3109/00498254.2011.645517] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The ways, how to increase effectiveness of doxorubicin (DOX) in cancer cells and decrease its toxicity in normal cells, have been intensively studied. In breast cancer cells MCF-7, isoquinoline derivative oracin (ORC) inhibited DOX reduction and increased DOX antiproliferative effect. The aim of this study was to test the influence of ORC on the reduction of DOX and its toxicity in hepatocytes and non-tumourous breast cells. The kinetics of DOX reduction was measured in cytosols from rat liver, human liver and human mammary epithelial cells MCF-10A. Activity and expression of carbonyl reductase 1 (CBR1) were assayed using menadione as a substrate and western blot analysis. End-point tests of viability served for study of cytotoxicity of DOX, ORC and DOX+ORC combinations in rat hepatocytes and MCF-10A cells. The inhibitory effect of ORC on DOX reductases was almost none in MCF-10A cells and mild in liver. CBR1 expression and activity was lower in non-tumourous MCF-10A cells than in cancer MCF-7 cells. Cytotoxicity tests showed that DOX+ORC combinations had significantly lower toxicity than DOX alone in MCF-10A cells as well as in hepatocytes. ORC significantly decreases DOX toxicity in MCF-10A and in hepatocytes. Therefore, concomitant use of ORC and DOX may protect normal cells against DOX toxicity.
Collapse
Affiliation(s)
- V Hanušová
- Department of Biochemical Sciences, Charles University, Hradec Králové, Czech Republic
| | | | | | | |
Collapse
|
215
|
Nawara K, Krysinski P, Blanchard GJ. Doxorubicin is a photocatalyst for the generation of H2O2. RSC Adv 2012. [DOI: 10.1039/c2ra20323e] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
216
|
|
217
|
Gilliam LAA, St Clair DK. Chemotherapy-induced weakness and fatigue in skeletal muscle: the role of oxidative stress. Antioxid Redox Signal 2011; 15:2543-63. [PMID: 21457105 PMCID: PMC3176345 DOI: 10.1089/ars.2011.3965] [Citation(s) in RCA: 224] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
SIGNIFICANCE Fatigue is one of the most common symptoms of cancer and its treatment, manifested in the clinic through weakness and exercise intolerance. These side effects not only compromise patient's quality of life (QOL), but also diminish physical activity, resulting in limited treatment and increased morbidity. RECENT ADVANCES Oxidative stress, mediated by cancer or chemotherapeutic agents, is an underlying mechanism of the drug-induced toxicity. Nontargeted tissues, such as striated muscle, are severely affected by oxidative stress during chemotherapy, leading to toxicity and dysfunction. CRITICAL ISSUES These findings highlight the importance of investigating clinically applicable interventions to alleviate the debilitating side effects. This article discusses the clinically available chemotherapy drugs that cause fatigue and oxidative stress in cancer patients, with an in-depth focus on the anthracycline doxorubicin. Doxorubicin, an effective anticancer drug, is a primary example of how chemotherapeutic agents disrupt striated muscle function through oxidative stress. FUTURE DIRECTIONS Further research investigating antioxidants could provide relief for cancer patients from debilitating muscle weakness, leading to improved quality of life.
Collapse
|
218
|
Gilliam LAA, Moylan JS, Patterson EW, Smith JD, Wilson AS, Rabbani Z, Reid MB. Doxorubicin acts via mitochondrial ROS to stimulate catabolism in C2C12 myotubes. Am J Physiol Cell Physiol 2011; 302:C195-202. [PMID: 21940668 DOI: 10.1152/ajpcell.00217.2011] [Citation(s) in RCA: 104] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Doxorubicin, a commonly prescribed chemotherapeutic agent, causes skeletal muscle wasting in cancer patients undergoing treatment and increases mitochondrial reactive oxygen species (ROS) production. ROS stimulate protein degradation in muscle by activating proteolytic systems that include caspase-3 and the ubiquitin-proteasome pathway. We hypothesized that doxorubicin causes skeletal muscle catabolism through ROS, causing upregulation of E3 ubiquitin ligases and caspase-3. We tested this hypothesis by exposing differentiated C2C12 myotubes to doxorubicin (0.2 μM). Doxorubicin decreased myotube width 48 h following exposure, along with a 40-50% reduction in myosin and sarcomeric actin. Cytosolic oxidant activity was elevated in myotubes 2 h following doxorubicin exposure. This increase in oxidants was followed by an increase in the E3 ubiquitin ligase atrogin-1/muscle atrophy F-box (MAFbx) and caspase-3. Treating myotubes with SS31 (opposes mitochondrial ROS) inhibited expression of ROS-sensitive atrogin-1/MAFbx and protected against doxorubicin-stimulated catabolism. These findings suggest doxorubicin acts via mitochondrial ROS to stimulate myotube atrophy.
Collapse
Affiliation(s)
- Laura A A Gilliam
- Department of Physiology, University of Kentucky, Lexington, Kentucky 40536-0298, USA
| | | | | | | | | | | | | |
Collapse
|
219
|
Mukherjee S, Das P, Das S. Exploration of small hydroxy-9,10-anthraquinones as anthracycline analogues: physicochemical characteristics and DNA binding for comparison. J PHYS ORG CHEM 2011. [DOI: 10.1002/poc.1928] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
| | - Piyal Das
- Department of Chemistry; Jadavpur University; Kolkata; 700032; India
| | - Saurabh Das
- Department of Chemistry; Jadavpur University; Kolkata; 700032; India
| |
Collapse
|
220
|
A switching mechanism in doxorubicin bioactivation can be exploited to control doxorubicin toxicity. PLoS Comput Biol 2011; 7:e1002151. [PMID: 21935349 PMCID: PMC3174179 DOI: 10.1371/journal.pcbi.1002151] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Accepted: 06/21/2011] [Indexed: 02/07/2023] Open
Abstract
Although doxorubicin toxicity in cancer cells is multifactorial, the enzymatic bioactivation of the drug can significantly contribute to its cytotoxicity. Previous research has identified most of the components that comprise the doxorubicin bioactivation network; however, adaptation of the network to changes in doxorubicin treatment or to patient-specific changes in network components is much less understood. To investigate the properties of the coupled reduction/oxidation reactions of the doxorubicin bioactivation network, we analyzed metabolic differences between two patient-derived acute lymphoblastic leukemia (ALL) cell lines exhibiting varied doxorubicin sensitivities. We developed computational models that accurately predicted doxorubicin bioactivation in both ALL cell lines at high and low doxorubicin concentrations. Oxygen-dependent redox cycling promoted superoxide accumulation while NADPH-dependent reductive conversion promoted semiquinone doxorubicin. This fundamental switch in control is observed between doxorubicin sensitive and insensitive ALL cells and between high and low doxorubicin concentrations. We demonstrate that pharmacological intervention strategies can be employed to either enhance or impede doxorubicin cytotoxicity in ALL cells due to the switching that occurs between oxygen-dependent superoxide generation and NADPH-dependent doxorubicin semiquinone formation. In the United States, acute lymphoblastic leukemia (ALL) is the most common form of cancer among children. Although the survival rate of childhood leukemia is relatively high, those who do not respond to chemotherapy have very low prognostic outcome. Recent reports point to the critical role of metabolism in determining cell sensitivity to doxorubicin, a conventional drug used in leukemia treatment. Most of the molecular components involved in doxorubicin metabolism have been identified; however, how these components operate as a system and how adaptation of the doxorubicin metabolic network to patient-specific changes in protein components is much less understood. We have therefore chosen to investigate via computational modeling the variations in the distribution of proteins that metabolize doxorubicin can control a cell's ability to respond to doxorubicin treatment. This systems-level approach provides a framework for understanding how patient-specific variability leads to patient-sensitivity to doxorubicin treatment at different doses. With this knowledge, we were able to correctly predict complex behavior induced by pharmacological intervention strategies for manipulation of doxorubicin metabolism. When our interventions are used in combination with doxorubicin, cell viability was promoted or potentiated based on dominant control mechanisms within the metabolic network.
Collapse
|
221
|
Zhu SG, Kukreja RC, Das A, Chen Q, Lesnefsky EJ, Xi L. Dietary nitrate supplementation protects against Doxorubicin-induced cardiomyopathy by improving mitochondrial function. J Am Coll Cardiol 2011; 57:2181-9. [PMID: 21596234 DOI: 10.1016/j.jacc.2011.01.024] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2010] [Revised: 01/07/2011] [Accepted: 01/11/2011] [Indexed: 11/29/2022]
Abstract
OBJECTIVES The aim of this study was to test the hypothesis that long-term dietary nitrate supplementation protects against doxorubicin-induced cardiomyopathy by improving ventricular function and reducing mitochondrial respiratory chain damage. BACKGROUND Doxorubicin is a powerful anthracycline antibiotic used to treat divergent human neoplasms. Its clinical use is limited because of severe cardiotoxic side effects. Dietary nitrate and nitrite are essential nutrients for maintenance of steady-state tissue levels of nitric oxide and may play a therapeutic role in diseases associated with nitric oxide insufficiency or dysregulation. Dietary nitrate and nitrite supplementation alleviates myocardial injury caused by ischemia-reperfusion and cardiac arrest-resuscitation. METHODS Adult male CF-1 mice were given a single dose of doxorubicin (15 mg/kg intraperitoneally), and left ventricular contractile function was assessed 5 days later using both echocardiography and pressure-volume Millar catheterization. A nitrate supplementation regimen (1 g/l sodium nitrate in drinking water) was started 7 days before doxorubicin injection and continued thereafter. Cardiomyocyte necrosis and apoptosis, tissue lipid peroxidation, and plasma nitrate and nitrite levels were assessed. In addition, mitochondrial complex I activity, oxidative phosphorylation capacity, and hydrogen peroxide generation were determined in parallel experiments. RESULTS Doxorubicin caused impairment of ventricular contractility and cell death, which were significantly reduced by nitrate supplementation (p < 0.05). These cardioprotective effects were associated with a significant decrease in tissue lipid peroxidation. Nitrate supplementation significantly preserved mitochondrial complex I activity and oxidative phosphorylation and attenuated hydrogen peroxide generation after doxorubicin treatment. CONCLUSIONS Long-term oral intake of inorganic nitrate attenuates doxorubicin-induced ventricular dysfunction, cell death, oxidative stress, and mitochondrial respiratory chain damage. Nitrate could be a promising therapeutic agent against doxorubicin-induced cardiotoxicity.
Collapse
Affiliation(s)
- Shu-Guang Zhu
- VCU Pauley Heart Center, Division of Cardiology, Virginia Commonwealth University, Richmond, Virginia 23298-0204, USA
| | | | | | | | | | | |
Collapse
|
222
|
Yao CX, Li WY, Zhang SF, Zhang SF, Zhang HF, Zang MX. Effects of Doxorubicin and Fenofibrate on the activities of NADH oxidase and citrate synthase in mice. Basic Clin Pharmacol Toxicol 2011; 109:452-6. [PMID: 21711451 DOI: 10.1111/j.1742-7843.2011.00748.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Doxorubicin (Dox) has widely been used as an anticancer drug, but its use is limited by serious toxicity to the heart, kidney and liver. Mitochondrial dysfunction is one of the potential mechanisms of toxicity but not fully understood. Fenofibrate, one of the peroxisome proliferator-activated receptor-alpha (PPARα) ligands, is involved in lipid metabolism which takes place primarily in the mitochondria, so mitochondrial function may be affected by fenofibrate. Therefore, we investigated the effects of DOX and fenofibrate on activities of both mitochondrial citrate synthase and NADH oxidase, which are marker enzymes in the tricarboxylic acid (TCA) cycle and a measure of the complex I-III-IV activity in electron transport chain, respectively. Dox (15 mg/kg) and/or fenofibrate (100 mg/kg/day) were administered to mice for 3 or 14 days, and the activities of citrate synthase and NADH oxidase were measured. Our study showed that Dox significantly inhibits the activity of citrate synthase while fenofibrate induces the activity. Similar to citrate synthase, NADH oxidase activity was also induced by fenofibrate except in spleen but inhibited by Dox except in the heart and liver. Furthermore, fenofibrate not only protects citrate synthase activity from Dox-induced toxicity in the ventricle but also significantly rescues NADH oxidase activity in the kidney. These results reveal the actions of fenofibrate and Dox on the mitochondria, and the underlying mechanism may be related to the toxicity of Dox, which has clinical implications in the side effects of Dox treatment by modulation of mitochondrial function.
Collapse
Affiliation(s)
- Chun-Xia Yao
- Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Henan, China
| | | | | | | | | | | |
Collapse
|
223
|
Peroxiredoxins are involved in metallothionein protection from doxorubicin cardiotoxicity. Eur J Pharmacol 2011; 659:224-32. [DOI: 10.1016/j.ejphar.2011.03.031] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2010] [Revised: 02/28/2011] [Accepted: 03/21/2011] [Indexed: 11/17/2022]
|
224
|
Ascensão A, Oliveira PJ, Magalhães J. Exercise as a beneficial adjunct therapy during Doxorubicin treatment--role of mitochondria in cardioprotection. Int J Cardiol 2011; 156:4-10. [PMID: 21636148 DOI: 10.1016/j.ijcard.2011.05.060] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Revised: 03/14/2011] [Accepted: 05/13/2011] [Indexed: 01/01/2023]
Abstract
One of the mostly used chemotherapeutic drugs is the highly effective anthracycline Doxorubicin. However, its clinical use is limited by the dose-related and cumulative cardiotoxicity and consequent dysfunction. It has been proposed that the etiology of this toxicity is related to mitochondrial dysfunction. The present review aimed to analyze the promising results regarding the effect of several types of physical exercise in cardiac tolerance of animals treated with acute and sub-chronic doses of Doxorubicin (DOX), highlighting the importance of cardiac mitochondrial-related mechanisms in the process. Physical exercise positively modulates some important cardiac defense systems to antagonize the toxic effects caused by DOX treatment, including antioxidant capacity, the overexpression of heat shock proteins and other anti-apoptotic proteins. An important role in this protective phenotype afforded by exercise should be attributed to mitochondrial plasticity, as related adaptations could be translated into improved cardiac function in the setting of the DOX cardiomyopathy.
Collapse
Affiliation(s)
- António Ascensão
- Research Centre in Physical Activity, Health and Leisure, Faculty of Sport, University of Porto, Portugal.
| | | | | |
Collapse
|
225
|
Štěrba M, Popelová O, Lenčo J, Fučíková A, Brčáková E, Mazurová Y, Jirkovský E, Šimůnek T, Adamcová M, Mičuda S, Stulík J, Geršl V. Proteomic insights into chronic anthracycline cardiotoxicity. J Mol Cell Cardiol 2011; 50:849-62. [DOI: 10.1016/j.yjmcc.2011.01.018] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2010] [Revised: 01/19/2011] [Accepted: 01/20/2011] [Indexed: 10/18/2022]
|
226
|
Kuznetsov AV, Margreiter R, Amberger A, Saks V, Grimm M. Changes in mitochondrial redox state, membrane potential and calcium precede mitochondrial dysfunction in doxorubicin-induced cell death. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2011; 1813:1144-52. [PMID: 21406203 DOI: 10.1016/j.bbamcr.2011.03.002] [Citation(s) in RCA: 148] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Revised: 02/09/2011] [Accepted: 03/03/2011] [Indexed: 10/18/2022]
Abstract
Mitochondria play central roles in cell life as a source of energy and in cell death by inducing apoptosis. Many important functions of mitochondria change in cancer, and these organelles can be a target of chemotherapy. The widely used anticancer drug doxorubicin (DOX) causes cell death, inhibition of cell cycle/proliferation and mitochondrial impairment. However, the mechanism of such impairment is not completely understood. In our study we used confocal and two-photon fluorescence imaging together with enzymatic and respirometric analysis to study short- and long-term effects of doxorubicin on mitochondria in various human carcinoma cells. We show that short-term (<30 min) effects include i) rapid changes in mitochondrial redox potentials towards a more oxidized state (flavoproteins and NADH), ii) mitochondrial depolarization, iii) elevated matrix calcium levels, and iv) mitochondrial ROS production, demonstrating a complex pattern of mitochondrial alterations. Significant inhibition of mitochondrial endogenous and uncoupled respiration, ATP depletion and changes in the activities of marker enzymes were observed after 48 h of DOX treatment (long-term effects) associated with cell cycle arrest and death.
Collapse
Affiliation(s)
- Andrey V Kuznetsov
- Department of Heart Surgery, Innsbruck Medical University, Innrain 66, Innsbruck A-6020, Austria.
| | | | | | | | | |
Collapse
|
227
|
Kruk I, Michalska T, Kładna A, Berczyński P, Aboul-Enein HY. Chemiluminescence investigations of antioxidative activities of some antibiotics against superoxide anion radical. LUMINESCENCE 2011; 26:598-603. [DOI: 10.1002/bio.1278] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2010] [Accepted: 01/04/2011] [Indexed: 11/10/2022]
Affiliation(s)
- Irena Kruk
- Institute of Physics; Faculty of Mechanical Engineering and Mechatronics; West Pomeranian University of Technology in Szczecin; Poland
| | - Teresa Michalska
- Institute of Physics; Faculty of Mechanical Engineering and Mechatronics; West Pomeranian University of Technology in Szczecin; Poland
| | - Aleksandra Kładna
- Department of Medical History and Ethics; Pomeranian Medical University of Szczecin; Poland
| | - Paweł Berczyński
- Institute of Physics; Faculty of Mechanical Engineering and Mechatronics; West Pomeranian University of Technology in Szczecin; Poland
| | - Hassan Y. Aboul-Enein
- Pharmaceutical and Medicinal Chemistry Department; Pharmacetical and Drug Industries Research Division; National Research Centre; Dokki; Cairo; 12311; Egypt
| |
Collapse
|
228
|
Kumar SN, Konorev EA, Aggarwal D, Kalyanaraman B. Analysis of proteome changes in doxorubicin-treated adult rat cardiomyocyte. J Proteomics 2011; 74:683-97. [PMID: 21338723 DOI: 10.1016/j.jprot.2011.02.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2010] [Revised: 02/10/2011] [Accepted: 02/12/2011] [Indexed: 12/27/2022]
Abstract
Doxorubicin-induced cardiomyopathy in cancer patients is well established. The proposed mechanism of cardiac damage includes generation of reactive oxygen species, mitochondrial dysfunction and cardiomyocyte apoptosis. Exposure of adult rat cardiomyocytes to low levels of DOX for 48h induced apoptosis. Analysis of protein expression showed a differential regulation of several key proteins including the voltage dependent anion selective channel protein 2 and methylmalonate semialdehyde dehydrogenase. In comparison, proteomic evaluation of DOX-treated rat heart showed a slightly different set of protein changes that suggests nuclear accumulation of DOX. Using a new solubilization technique, changes in low abundant protein profiles were monitored. Altered protein expression, modification and function related to oxidative stress response may play an important role in DOX cardiotoxicity.
Collapse
Affiliation(s)
- Suresh N Kumar
- Department of Pathology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | | | | | | |
Collapse
|
229
|
Dursun N, Taşkın E, Yerer Aycan MB, Şahin L. Selenium-mediated cardioprotection against adriamycin-induced mitochondrial damage. Drug Chem Toxicol 2011; 34:199-207. [DOI: 10.3109/01480545.2010.538693] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
230
|
Das P, Guin PS, Mandal PC, Paul M, Paul S, Das S. Cyclic voltammetric studies of 1,2,4-trihydroxy-9,10-anthraquinone, its interaction with calf thymus DNA and anti-leukemic activity on MOLT-4 cell lines: a comparison with anthracycline anticancer drugs. J PHYS ORG CHEM 2011. [DOI: 10.1002/poc.1827] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
231
|
Gilliam LAA, Moylan JS, Callahan LA, Sumandea MP, Reid MB. Doxorubicin causes diaphragm weakness in murine models of cancer chemotherapy. Muscle Nerve 2011; 43:94-102. [PMID: 21171100 DOI: 10.1002/mus.21809] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Doxorubicin is a chemotherapeutic agent prescribed for a variety of tumors. While undergoing treatment, patients exhibit frequent symptoms that suggest respiratory muscle weakness. Cancer patients can receive doxorubicin chemotherapy through either intravenous (IV) or intraperitoneal (IP) injections. We hypothesized that respiratory muscle function would be depressed in a murine model of chemotherapy. We tested this hypothesis by treating C57BL/6 mice with a clinical dose of doxorubicin (20 mg/kg) via IV or IP injection. Three days later we measured contractile properties of muscle fiber bundles isolated from the diaphragm. Doxorubicin consistently depressed diaphragm force with both methods of administration (P < 0.01). Doxorubicin IP exaggerated the depression in diaphragm force and stimulated tissue inflammation and muscle fiber injury. These results suggest that clinically relevant doses of doxorubicin cause respiratory muscle weakness and that the loss of function depends, in part, on the route of administration.
Collapse
Affiliation(s)
- Laura A A Gilliam
- Department of Physiology, University of Kentucky, 800 Rose Street, MS-508, Lexington, Kentucky 40536-0298, USA
| | | | | | | | | |
Collapse
|
232
|
Volkova M, Palmeri M, Russell KS, Russell RR. Activation of the aryl hydrocarbon receptor by doxorubicin mediates cytoprotective effects in the heart. Cardiovasc Res 2011; 90:305-14. [PMID: 21233252 DOI: 10.1093/cvr/cvr007] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
AIMS Doxorubicin (DOX) is a highly effective chemotherapeutic agent; however, cumulative dose-dependent cardiotoxicity is a significant side effect of this therapy. Because DOX is a polyaromatic hydrocarbon, we hypothesized that it will be metabolized by the activation of the aryl hydrocarbon receptor (AhR), a ligand-activated transcription factor that is involved in the metabolism of numerous xenobiotic agents. These studies were performed to determine whether DOX activates AhR and whether this activation modulates the toxicity of DOX in cardiomyocytes. METHODS AND RESULTS Treatment with DOX induced AhR migration to the nucleus, increased AhR binding with its co-factor, aryl hydrocarbon receptor nuclear translocator-1 (ARNT1), and increased the expression of AhR-regulated phase I (CYP1A1) and phase II (GSTA1) drug-metabolizing enzymes in both cardiomyocytes and in the intact heart. Knockdown of AhR in H9C2 cells abolished DOX-induced increases in CYP1A1 and GSTA1 expression. Similar results were obtained by treating adult rat ventricular myocytes with the AhR antagonist, CH-223191. Taken together, these findings indicate that DOX-induced upregulation of CYP1A1 and GSTA1 expression is AhR dependent. AhR null mice treated with 10 mg/kg DOX did not show any activation of CYP1A1 or GSTA1 expression. Moreover, lack of AhR in vivo resulted in a significant decrease in left ventricular function compared with wild-type animals, and increased p53 activation and apoptosis in the heart after treatment with DOX. CONCLUSIONS These findings indicate that AhR plays an important role in DOX metabolism by the heart and further demonstrate that AhR is cardioprotective against DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Maria Volkova
- Section of Cardiovascular Medicine, Yale University School of Medicine, 333 Cedar Street, FMP 3, New Haven, CT 06520, USA
| | | | | | | |
Collapse
|
233
|
Torres VM, Srdjenovic B, Jacevic V, Simic VD, Djordjevic A, Simplício AL. Fullerenol C60(OH)24 prevents doxorubicin-induced acute cardiotoxicity in rats. Pharmacol Rep 2011; 62:707-18. [PMID: 20885011 DOI: 10.1016/s1734-1140(10)70328-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2009] [Revised: 12/08/2009] [Indexed: 12/21/2022]
Abstract
Results obtained in vitro suggested that fullerenol's antiproliferative properties and protective effects against doxorubicin (DOX) cytotoxicity are mediated by antioxidative and hydroxyl radical scavenger activity. The aim of this study was to examine the influence of fullerenol on acute cardiotoxicity after the administration of a single high dose of DOX in vivo. The experiment was performed on male Wistar rats randomly divided into five groups, each containing eight individuals, that were treated as follows: I) 0.9% NaCl, II) 10 mg/kg DOX, III) 50 mg/kg fullerenol 30 min before 10 mg/kg DOX, IV) 100 mg/kg fullerenol 30 min before 10 mg/kg DOX, and V) 100 mg/kg fullerenol. A functional, biochemical, hematological, and pathomorphological examination of the heart as well as an evaluation of oxidative stress parameters was conducted on days 2 and 14 after DOX administration. The function of the heart was investigated by monitoring heart contractility after the adrenaline infusion. Fullerenol, applied alone, did not alter basal values of investigated animals. Both doses of fullerenol, used as a pretreatment, did not alter the basal parameters of the animals. The 100 mg/kg dose of fullerenol showed better protection. Considering the mechanisms of DOX toxicity, fullerenol likely exerts its protective role as a free radical sponge and/or by removing free iron through the formation of a fullerenol-iron complex. Our results suggest that fullerenol might be a potential cardioprotective agent in DOX-treated individuals.
Collapse
Affiliation(s)
- Vukosava Milic Torres
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Av. da República, Estação Agronómica Nacional, 2780-157 Oeiras, Portugal.
| | | | | | | | | | | |
Collapse
|
234
|
Guin PS, Das S, Mandal PC. Electrochemical Reduction of Quinones in Different Media: A Review. INTERNATIONAL JOURNAL OF ELECTROCHEMISTRY 2011. [DOI: 10.4061/2011/816202] [Citation(s) in RCA: 262] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The electron transfer reactions involving quinones, hydroquinones, and catechols are very important in many areas of chemistry, especially in biological systems. The therapeutic efficiency as well as toxicity of anthracycline anticancer drugs, a class of anthraquinones, is governed by their electrochemical properties. Other quinones serve as important functional moiety in various biological systems like electron-proton carriers in the respiratory chain and their involvement in photosynthetic electron flow systems. The present paper summarizes literatures on the reduction of quinones in different solvents under various conditions using different electrochemical methods. The influence of different reaction conditions including pH of the media, nature of supporting electrolytes, nature of other additives, intramolecular or intermolecular hydrogen bonding, ion pair formation, polarity of the solvents, stabilization of the semiquinone and quinone dianion, catalytic property, and adsorption at the electrode surface, are discussed and relationships between reaction conditions and products formed have been presented.
Collapse
Affiliation(s)
- Partha Sarathi Guin
- Department of Chemistry, Shibpur Dinobundhoo Institution (College), 412/1 G. T. Road (South), Howrah 711102, India
| | - Saurabh Das
- Department of Chemistry, Jadavpur University, Raja S. C. Mullick Road, Kolkata 700032, India
| | - P. C. Mandal
- Chemical Sciences Division, Saha Institute of Nuclear Physics, 1/AF-Bidhannagar, Kolkata 700064, India
| |
Collapse
|
235
|
Mukhopadhyay P, Horváth B, Rajesh M, Matsumoto S, Saito K, Bátkai S, Patel V, Tanchian G, Gao RY, Cravatt BF, Haskó G, Pacher P. Fatty acid amide hydrolase is a key regulator of endocannabinoid-induced myocardial tissue injury. Free Radic Biol Med 2011; 50:179-95. [PMID: 21070851 PMCID: PMC3022384 DOI: 10.1016/j.freeradbiomed.2010.11.002] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2010] [Revised: 10/28/2010] [Accepted: 11/01/2010] [Indexed: 12/22/2022]
Abstract
Previous studies have suggested that increased levels of endocannabinoids in various cardiovascular disorders (e.g., various forms of shock, cardiomyopathies, atherosclerosis) through the activation of CB(1) cannabinoid receptors may promote cardiovascular dysfunction and tissue injury. We have investigated the role of the main endocannabinoid anandamide-metabolizing enzyme (fatty acid amide hydrolase; FAAH) in myocardial injury induced by an important chemotherapeutic drug, doxorubicin (DOX; known for its cardiotoxicity mediated by increased reactive oxygen and nitrogen species generation), using well-established acute and chronic cardiomyopathy models in mice. The DOX-induced myocardial oxidative/nitrative stress (increased 4-hydroxynonenal, protein carbonyl, and nitrotyrosine levels and decreased glutathione content) correlated with multiple cell death markers, which were enhanced in FAAH knockout mice exhibiting significantly increased DOX-induced mortality and cardiac dysfunction compared to their wild type. The effects of DOX in FAAH knockouts were attenuated by CB(1) receptor antagonists. Furthermore, anandamide induced enhanced cell death in human cardiomyocytes pretreated with FAAH inhibitor and enhanced sensitivity to ROS generation in inflammatory cells of FAAH knockouts. These results suggest that in pathological conditions associated with acute oxidative/nitrative stress FAAH plays a key role in controlling the tissue injury that is, at least in part, mediated by the activation of CB(1) receptors by endocannabinoids.
Collapse
MESH Headings
- Amidohydrolases/genetics
- Amidohydrolases/metabolism
- Amidohydrolases/physiology
- Animals
- Antineoplastic Agents/adverse effects
- Cannabinoid Receptor Modulators/adverse effects
- Cannabinoid Receptor Modulators/pharmacology
- Cardiomyopathies/chemically induced
- Cardiomyopathies/genetics
- Cardiomyopathies/metabolism
- Cardiomyopathies/pathology
- Cells, Cultured
- Doxorubicin/adverse effects
- Endocannabinoids
- Heart/drug effects
- Humans
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Myocardium/metabolism
- Myocardium/pathology
- Reactive Oxygen Species/metabolism
- Receptor, Cannabinoid, CB1/agonists
- Receptor, Cannabinoid, CB1/antagonists & inhibitors
- Receptor, Cannabinoid, CB1/metabolism
- Receptor, Cannabinoid, CB1/physiology
Collapse
Affiliation(s)
- Partha Mukhopadhyay
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
| | - Bėla Horváth
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
| | - Mohanraj Rajesh
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
| | - Shingo Matsumoto
- Radiation Biology Branch, NCI, National Institutes of Health, Bethesda, Maryland, USA
| | - Keita Saito
- Radiation Biology Branch, NCI, National Institutes of Health, Bethesda, Maryland, USA
| | - Sándor Bátkai
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
| | - Vivek Patel
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
| | - Galin Tanchian
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
| | - Rachel Y Gao
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
| | - Benjamin F. Cravatt
- The Skaggs Institute for Chemical Biology and Department of Cell Biology, The Scripps Research Institute, La Jolla, California
| | - György Haskó
- Department of Surgery, University of Medicine and Dentistry of New Jersey-New Jersey Medical School, Newark, New Jersey 07103, USA
| | - Pál Pacher
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
236
|
Arechederra MN, Addo PK, Minteer SD. Poly(neutral red) as a NAD+ reduction catalyst and a NADH oxidation catalyst: Towards the development of a rechargeable biobattery. Electrochim Acta 2011. [DOI: 10.1016/j.electacta.2010.10.045] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
237
|
Acute exercise protects against calcium-induced cardiac mitochondrial permeability transition pore opening in doxorubicin-treated rats. Clin Sci (Lond) 2010; 120:37-49. [PMID: 20666733 DOI: 10.1042/cs20100254] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The use of DOX (doxorubicin), an antibiotic used in oncological treatments, is limited by a dose-related cardiotoxicity against which acute exercise is protective. However, the mitochondrial-related mechanisms of this protection remain unknown. Therefore the present study aimed to determine the effects of an acute endurance exercise bout performed 24 h before DOX treatment on heart and liver mitochondrial function. A total of 20 adult male Wistar rats were divided into groups as follows: non-exercised with saline (NE + SAL), non-exercised DOX-treated (NE + DOX), exercised with saline (EX + SAL) and exercised DOX-treated (EX + DOX). The animals performed a 60 min exercise bout on a treadmill or remained sedentary 24 h before receiving either a DOX bolus (20 mg/kg of body weight) or saline. Heart and liver mitochondrial function [oxygen consumption, membrane potential (DeltaPsi) and cyclosporin-A-sensitive calcium-induced MPTP (mitochondrial permeability transition pore) opening] were evaluated. The activities of the respiratory complex, Mn-SOD (superoxide dismutase), caspases 3 and 9, as well as the levels of ANT (adenine nucleotide translocase), VDAC (voltage-dependent anion channel), CypD (cyclophilin D), Bax and Bcl-2, were measured. Acute exercise prevented the decreased cardiac mitochondrial function (state 3, phosphorylative lagphase; maximal DeltaPsi generated both with complex I- and II-linked substrates and calcium-induced MPTP opening) induced by DOX treatment. Exercise also prevented the DOX-induced decreased activity of cardiac mitochondrial chain complexes I and V, and increased caspase 3 and 9 activities. DOX administration and exercise caused increased cardiac mitochondrial SOD activity. Exercise ameliorated liver mitochondrial complex activities. No alterations were observed in the measured MPTP and apoptosis-related proteins in heart and liver mitochondria. The results demonstrate that acute exercise protects against cardiac mitochondrial dysfunction, preserving mitochondrial phosphorylation capacity and attenuating DOX-induced decreased tolerance to MPTP opening.
Collapse
|
238
|
Shuai Y, Guo J, Dong Y, Zhong W, Xiao P, Zhou T, Zhang L, Peng S. Global gene expression profiles of MT knockout and wild-type mice in the condition of doxorubicin-induced cardiomyopathy. Toxicol Lett 2010; 200:77-87. [PMID: 21040762 DOI: 10.1016/j.toxlet.2010.10.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2010] [Revised: 10/22/2010] [Accepted: 10/25/2010] [Indexed: 10/18/2022]
Abstract
Increasing evidence from in vivo and in vitro studies has indicated that MT exerts protective effects against DOX-induced cardiotoxicity; however the underlying precise mechanisms still remain an enigma. Therefore, the present study was designed using MT knockout mice in concert with genomic approaches to explore the possible molecular and cellular mechanisms in terms of the genetic network changes. MT-I/II null (MT⁻/⁻) mice and corresponding wild-type mice (MT+/+) were administrated with a single dose of DOX (15 mg/kg, i.p.) or equal volume of saline. Animals were sacrificed on the 4th day after DOX administration and samples were collected for further analyses. Global gene expression profiles of cardiac mRNA from two genotype mice revealed that 381 characteristically MT-responsive genes were identified between MT+/+ mice and MT⁻/⁻ mice in response to DOX, including fos, ucp3, car3, atf3, map3k6, etc. Functional analysis implied MAPK signaling pathway, p53 signaling pathway, Jak-STAT signaling pathway, PPAR signaling pathway, Wnt signaling pathway, etc. might be involved to mediate the protection of DOX cardiomyopathy by MT. Results from the present study not only validated the previously reported possible mechanisms of MT protection against DOX toxicity, but also provided new clues into the molecular mechanisms involved in this process.
Collapse
Affiliation(s)
- Yi Shuai
- Evaluation and Research Center for Toxicology, Institute of Disease Control and Prevention, Academy of Military Medical Science, 20 Dongdajie Street, Fengtai District, Beijing 100071, PR China
| | | | | | | | | | | | | | | |
Collapse
|
239
|
Al Samri MT, Al Shamsi M, Al-Salam S, Marzouqi F, Al Mansouri A, Al-Hammadi S, Balhaj G, Al Dawaar SKM, Al Hanjeri RSMS, Benedict S, Sudhadevi M, Conca W, Penefsky HS, Souid AK. Measurement of oxygen consumption by murine tissues in vitro. J Pharmacol Toxicol Methods 2010; 63:196-204. [PMID: 21034836 DOI: 10.1016/j.vascn.2010.10.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2010] [Revised: 10/16/2010] [Accepted: 10/19/2010] [Indexed: 10/18/2022]
Abstract
INTRODUCTION A novel in vitro system was developed to measure O₂ consumption by murine tissues over several hours. METHODS Tissue specimens (7-35 mg) excised from male Balb/c mice were immediately immersed in ice-cold Krebs-Henseleit buffer, saturated with 95% O₂:5% CO₂. The specimens were incubated at 37 °C in the buffer, continuously gassed with O₂:CO₂ (95:5). [O₂] was determined as a function of time from the phosphorescence decay rates (1/τ) of Pd(II) meso-tetra-(4-sulfonatophenyl)-tetrabenzoporphyrin. The values of 1/τ were linear with [O₂]: 1/τ=1/τo + kq [O₂]; 1/τo=the decay rate for zero O₂, kq=the rate constant in s⁻¹ μM⁻¹. RESULTS NaCN inhibited O₂ consumption, confirming oxidation occurred in the mitochondrial respiratory chain. The rate of respiration in lung specimens incubated in vitro for 3.9≤t≤12.4 h was 0.24±0.03 μM O₂ min⁻¹ mg⁻¹ (mean±SD, n=28). The corresponding rate for the liver was 0.27±0.13 (n=11, t≤4.7 h), spleen 0.28± 0.07 (n=10, t≤5h), kidney 0.34±0.12 (n=7, t≤5h) and pancreas 0.35±0.09 (n=10, t≤4h). Normal tissue histology at hour 5 was confirmed by light and electron microscopy. There was negligible number of apoptotic cells by caspase 3 staining. DISCUSSION This approach allows accurate assessment of tissue bioenergetics in vitro.
Collapse
Affiliation(s)
- Mohammed T Al Samri
- Department of Pediatrics, Faculty of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates 17666, UAE
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
240
|
Doxorubicin in vivo rapidly alters expression and translation of myocardial electron transport chain genes, leads to ATP loss and caspase 3 activation. PLoS One 2010; 5:e12733. [PMID: 20856801 PMCID: PMC2939875 DOI: 10.1371/journal.pone.0012733] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2010] [Accepted: 07/14/2010] [Indexed: 01/08/2023] Open
Abstract
Background Doxorubicin is one of the most effective anti-cancer drugs but its use is limited by cumulative cardiotoxicity that restricts lifetime dose. Redox damage is one of the most accepted mechanisms of toxicity, but not fully substantiated. Moreover doxorubicin is not an efficient redox cycling compound due to its low redox potential. Here we used genomic and chemical systems approaches in vivo to investigate the mechanisms of doxorubicin cardiotoxicity, and specifically test the hypothesis of redox cycling mediated cardiotoxicity. Methodology/Principal Findings Mice were treated with an acute dose of either doxorubicin (DOX) (15 mg/kg) or 2,3-dimethoxy-1,4-naphthoquinone (DMNQ) (25 mg/kg). DMNQ is a more efficient redox cycling agent than DOX but unlike DOX has limited ability to inhibit gene transcription and DNA replication. This allowed specific testing of the redox hypothesis for cardiotoxicity. An acute dose was used to avoid pathophysiological effects in the genomic analysis. However similar data were obtained with a chronic model, but are not specifically presented. All data are deposited in the Gene Expression Omnibus (GEO). Pathway and biochemical analysis of cardiac global gene transcription and mRNA translation data derived at time points from 5 min after an acute exposure in vivo showed a pronounced effect on electron transport chain activity. This led to loss of ATP, increased AMPK expression, mitochondrial genome amplification and activation of caspase 3. No data gathered with either compound indicated general redox damage, though site specific redox damage in mitochondria cannot be entirely discounted. Conclusions/Significance These data indicate the major mechanism of doxorubicin cardiotoxicity is via damage or inhibition of the electron transport chain and not general redox stress. There is a rapid response at transcriptional and translational level of many of the genes coding for proteins of the electron transport chain complexes. Still though ATP loss occurs with activation caspase 3 and these events probably account for the heart damage.
Collapse
|
241
|
Li J, Liu H, Ramachandran S, Waypa GB, Yin JJ, Li CQ, Han M, Huang HH, Sillard WW, Vanden Hoek TL, Shao ZH. Grape seed proanthocyanidins ameliorate Doxorubicin-induced cardiotoxicity. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2010; 38:569-84. [PMID: 20503473 DOI: 10.1142/s0192415x10008068] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Doxorubicin (Dox) is one of the most widely used and successful chemotherapeutic antitumor drugs. Its clinical application is highly limited due to its cumulative dose-related cardiotoxicity. Proposed mechanisms include the generation of reactive oxygen species (ROS)-mediated oxidative stress. Therefore, reducing oxidative stress should be protective against Dox-induced cardiotoxicity. To determine whether antioxidant, grape seed proanthocyanidin extract (GSPE) attenuates Dox-induced ROS generation and protects cardiomyocytes from Dox-induced oxidant injury, cultured primary cardiomyocytes were treated with doxorubicin (Dox, 10 microM) alone or GSPE (50 microg/ml) with Dox (10 microM) for 24 hours. Dox increased intracellular ROS production as measured by 6-carboxy-2',7'-dichlorodihydrofluorescein diacetate, induced significant cell death as assessed by propidium iodide, and declined the redox ratio of reduced glutathione (GSH)/oxidized glutathione (GSSG) and disrupted mitochondrial membrane potential as determined by 5,5',6,6'-tetrachloro-1,1',3,3'-tetraethlbenzimidazole-carbocyanide iodine (JC-1). Analysis of agarose gel electrophoresis revealed Dox-induced nuclear DNA damage with the ladder like fragmentation. GSPE treatment suppressed those alterations. Electron Spin Resonance (ESR) spectroscopy data also showed that GSPE strongly scavenged hydroxyl radical, superoxide and DPPH radicals. Together, these findings indicate that GSPE in combination with Dox has protective effect against Dox-induced toxicity in cardiomyocytes, which may be in part attributed to its antioxidative activity. Importantly, flow cytometric analysis demonstrated that co-treatment of Dox and GSPE did not decrease the proliferation-inhibitory effect of Dox in MCF-7 human breast carcinoma cells. Thus, GSPE may be a promising adjuvant to prevent cardiotoxicity without interfering with antineoplastic activity during chemotherapeutic treatment with Dox.
Collapse
Affiliation(s)
- Jing Li
- Emergency Resuscitation Center, Department of Medicine, University of Chicago, IL 60637, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
242
|
Lipid replacement therapy: a nutraceutical approach for reducing cancer-associated fatigue and the adverse effects of cancer therapy while restoring mitochondrial function. Cancer Metastasis Rev 2010; 29:543-52. [DOI: 10.1007/s10555-010-9245-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
243
|
Saalu L, Osinubi A, Oguntola J, Adeneye I, Benebo A. The Delayed Testicular Morphologic Effects of Doxorubicin and the
Rejuvinating Role of Grapefruit Seed Extract. INT J PHARMACOL 2010. [DOI: 10.3923/ijp.2010.192.199] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
244
|
Alkreathy H, Damanhouri ZA, Ahmed N, Slevin M, Ali SS, Osman AMM. Aged garlic extract protects against doxorubicin-induced cardiotoxicity in rats. Food Chem Toxicol 2010; 48:951-6. [DOI: 10.1016/j.fct.2010.01.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2009] [Revised: 12/26/2009] [Accepted: 01/05/2010] [Indexed: 10/20/2022]
|
245
|
Frias MA, Lang U, Gerber-Wicht C, James RW. Native and reconstituted HDL protect cardiomyocytes from doxorubicin-induced apoptosis. Cardiovasc Res 2010; 85:118-26. [PMID: 19700468 DOI: 10.1093/cvr/cvp289] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
AIMS We analysed the impact of native and reconstituted HDL on doxorubicin-induced cardiomyocyte apoptosis. While it is an effective anti-cancer agent, doxorubicin has serious cardiotoxic side effects. HDL has been shown to protect cardiomyocytes, notably against oxidative stress. METHODS AND RESULTS Cultured neonatal rat ventricular cardiomyocytes were subjected to doxorubicin-induced stress, monitored as caspase3 activation, apoptotic DNA fragmentation and cell viability. The protective effects of HDL and sphingosine-1-phosphate (S1P) were investigated using native HDL, reconstituted HDL of varied composition and agonists and antagonists of S1P receptors. Anti-apoptotic signalling pathways were identified with specific inhibitors. Native and reconstituted HDL significantly decreased doxorubicin-induced cardiomyocyte apoptosis, essentially due to the S1P component of HDL. The latter was mediated by the S1P2 receptor, but not the S1P1 or S1P3 receptors. The extracellular signal-regulated kinases 1 and 2 (ERK1/2) signalling pathway was required for the anti-apoptotic effects of HDL and S1P. The transcription factor Stat3 also played an important role, as inhibition of its activity compromised the protective effects of HDL and S1P on doxorubicin-induced apoptosis. CONCLUSION HDL and its sphingosine-1-phosphate component can protect cardiomyocytes against doxorubicin toxicity and may offer one means of reducing cardiotoxic side effects during doxorubicin therapy. The study identified anti-apoptotic pathways that could be exploited to improve cardiomyocyte survival.
Collapse
Affiliation(s)
- Miguel A Frias
- University of Geneva, Service of Endocrinology, Diabetology, and Nutrition, University Hospital, CH-1211 Geneva 14, Switzerland
| | | | | | | |
Collapse
|
246
|
Matsuura C, Brunini TM, Carvalho LC, Resende AC, Carvalho JJ, de Castro JPW, Mendes-Ribeiro AC. Exercise training in doxorubicin-induced heart failure: effects on the L-arginine–NO pathway and vascular reactivity. ACTA ACUST UNITED AC 2010; 4:7-13. [DOI: 10.1016/j.jash.2009.10.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2009] [Revised: 10/05/2009] [Accepted: 10/20/2009] [Indexed: 11/25/2022]
|
247
|
Saalu L, Osinubi A, Jewo P, Oyewopo A, Ajayi G. An Evaluation of Influence of Citrus paradisi Seed Extract on Doxorubicin-Induced Testicular Oxidative Stress and Impaired Spermatogenesis. ACTA ACUST UNITED AC 2009. [DOI: 10.3923/ajsr.2010.51.61] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
248
|
Valls-Belles V, Torres C, Muñiz P, Codoñer-Franch P. Effect of beer consumption on levels of complex I and complex IV liver and heart mitochondrial enzymes and coenzymes Q9 and Q10 in adriamycin-treated rats. Eur J Nutr 2009; 49:181-7. [PMID: 19841853 DOI: 10.1007/s00394-009-0064-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2009] [Accepted: 09/30/2009] [Indexed: 01/08/2023]
Abstract
BACKGROUND There is increasing evidence indicating that the dietary intake of food with high antioxidant capacity may protect mitochondria from damage and exert positive effects on different pathogenic processes. AIM OF THE STUDY The present study was designed to evaluate the possible protective effect of alcohol-free beer intake on chain components dysfunction of liver and heart mitochondria, and to compare with the effect of alcohol beer intake. METHODS The study was carried out in rat heart and liver mitochondria by inducing with Adriamycin the dysfunction of the respiratory chain. Heart and liver mitochondria were isolated from rats and subjected to oxidative stress with two doses of Adriamycin (5 mg/Kg) 7 days from the beginning of consumption of both alcohol-free and alcohol beer during 31 days. Complexes I and IV and the levels of coenzymes Q(9) and Q(10) were evaluated and compared with a control group. RESULTS Liver and heart mitochondria isolated from rats treated with Adryamicin showed a decrease in levels of complex I and complex IV enzymatic activity and in levels of coenzymes Q(9) and Q(10). Beer intake for itself does not affect any of the studied parameters. Therefore, the consumption of both alcohol and alcohol-free beer by rats treated with Adriamycin prevents the inhibition of enzymatic activities of complexes I and IV and the oxidation of coenzymes Q(9) and Q(10) in rat heart and liver mitochondria. CONCLUSIONS These results indicate that alcohol-free beer prevents adriamycin-induced damage to mitochondrial chain components and, therefore, helps to prevent mitochondrial dysfunction.
Collapse
Affiliation(s)
- Victoria Valls-Belles
- Departamento de Pediatría, Ginecología y Obstetricia, Universidad de Valencia, Spain.
| | | | | | | |
Collapse
|
249
|
Abstract
To clarify the mechanism of the cardiotoxic action of adriamycin (ADM), the participation of free radicals from ADM in cardiotoxicity was investigated through the protective action of glutathione (GSH) or by using electron spin resonance (ESR). Oxidation of ADM by horseradish peroxidase and H2O2 (HRP-H2O2) was blocked by GSH concentration dependently. Inactivation of creatine kinase (CK) induced during interaction of ADM with HRP-H2O2 was also protected by GSH. Other anthracycline antitumor drugs that have a p-hydroquinone structure in the B ring also inactivated CK, and GSH inhibited the inactivation of CK. These results suggest that ADM was activated through oxidation of the p-hydroquinone in the B ring by HRP-H2O2. Although ESR signals of the oxidative ADM B ring semiquinone were not detected, glutathionyl radicals were formed during the interaction of ADM with HRP-H2O2 in the presence of GSH. ADM may be oxidized to the ADM B ring semiquinone and then reacts with the SH group. However, ESR signals of ADM C ring semiquinone, which was reductively formed by xanthine oxidase (XO) and hypoxanthine (HX) under anaerobic conditions, were not diminished by GSH, but they completely disappeared with ferric ion. These results indicate that oxidative ADM B ring semiquinones oxidized the SH group in CK, but reductive ADM C ring semiquinone radicals may participate in the oxidation of lipids or DNA and not of the SH group.
Collapse
Affiliation(s)
- Sanae Muraoka
- Department of Biology, Hokkaido College of Pharmacy, Katsuraoka-cho 7-1, Otaru 0470264, Japan
| | | |
Collapse
|
250
|
Marczak A, Jóźwiak Z. Damage to the cell antioxidative system in human erythrocytes incubated with idarubicin and glutaraldehyde. Toxicol In Vitro 2009; 23:1188-94. [PMID: 19490936 DOI: 10.1016/j.tiv.2009.05.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2009] [Revised: 04/30/2009] [Accepted: 05/25/2009] [Indexed: 11/17/2022]
Abstract
Encapsulation of antineoplastic drugs within erythrocytes is one of the studied strategies to diminish the toxic side effects of anthracycline antibiotics. Glutaraldehyde is often used as crosslinking agent to link the drugs, including idarubicin (IDA) to the cells. The previous studies indicated that in glutaraldehyde-treated human erythrocytes the elevated level of drug was observed but also the various changes in the organization of the red cells were noted. In this study, we continue our investigations and now we concentrate on the effect of these compounds on antioxidative system in erythrocytes. We determined reactive oxygen species (ROS) production, glutathione content and alterations in the activity of enzymes responsible for maintaining glutathione in reduced form in human erythrocytes. Measurements of both reduced and total glutathione levels and the activity of glutathione reductase and glucose-6-phosphate dehydrogenase were performed spectrophotometrically. The results show that ROS were produced in erythrocytes treated with IDA and with IDA and glutaraldehyde. IDA at a concentration of 10 microg/ml did not cause any changes in total or reduced glutathione levels. When IDA-preincubated erythrocytes were treated with glutaraldehyde, significant changes in the determined parameters were observed in a glutaraldehyde concentration dependent manner. It was correlated with decreased activity of glutathione reductase (GR) and glucose-6-phosphate dehydrogenase (G6PD). Together with the significant changes in reduced form of glutathione (GSH)/total glutathione ratio, the exposure of phosphatidylserine at the cell surface was also observed.
Collapse
Affiliation(s)
- Agnieszka Marczak
- Department of Thermobiology, University of Łódź, 90-237 Łódź, Poland.
| | | |
Collapse
|