201
|
Singh P, Sharma R, McElhanon K, Allen CD, Megyesi JK, Beneš H, Singh SP. Sulforaphane protects the heart from doxorubicin-induced toxicity. Free Radic Biol Med 2015; 86:90-101. [PMID: 26025579 PMCID: PMC4554811 DOI: 10.1016/j.freeradbiomed.2015.05.028] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 05/04/2015] [Accepted: 05/19/2015] [Indexed: 01/08/2023]
Abstract
Cardiotoxicity is one of the major side effects encountered during cancer chemotherapy with doxorubicin (DOX) and other anthracyclines. Previous studies have shown that oxidative stress caused by DOX is one of the primary mechanisms for its toxic effects on the heart. Since the redox-sensitive transcription factor, Nrf2, plays a major role in protecting cells from the toxic metabolites generated during oxidative stress, we examined the effects of the phytochemical sulforaphane (SFN), a potent Nrf2-activating agent, on DOX-induced cardiotoxicity. These studies were carried out both in vitro and in vivo using rat H9c2 cardiomyoblast cells and wild type 129/sv mice, and involved SFN pretreatment followed by SFN administration during DOX exposure. SFN treatment protected H9c2 cells from DOX cytotoxicity and also resulted in restored cardiac function and a significant reduction in DOX-induced cardiomyopathy and mortality in mice. Specificity of SFN induction of Nrf2 and protection of H9c2 cells was demonstrated in Nrf2 knockdown experiments. Cardiac accumulation of 4-hydroxynonenal (4-HNE) protein adducts, due to lipid peroxidation following DOX-induced oxidative stress, was significantly attenuated by SFN treatment. The respiratory function of cardiac mitochondria isolated from mice exposed to DOX alone was repressed, while SFN treatment with DOX significantly elevated mitochondrial respiratory complex activities. Co-administration of SFN reversed the DOX-associated reduction in nuclear Nrf2 binding activity and restored cardiac expression of Nrf2-regulated genes at both the RNA and protein levels. Together, our results demonstrate for the first time that the Nrf2 inducer, SFN, has the potential to provide protection against DOX-mediated cardiotoxicity.
Collapse
Affiliation(s)
- Preeti Singh
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| | - Rajendra Sharma
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Kevin McElhanon
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| | - Charles D Allen
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| | - Judit K Megyesi
- Central Arkansas Veterans Healthcare System, Little Rock, AR, USA; Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Helen Beneš
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Sharda P Singh
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; Central Arkansas Veterans Healthcare System, Little Rock, AR, USA.
| |
Collapse
|
202
|
Wang Y, Mei X, Yuan J, Lu W, Li B, Xu D. Taurine zinc solid dispersions attenuate doxorubicin-induced hepatotoxicity and cardiotoxicity in rats. Toxicol Appl Pharmacol 2015; 289:1-11. [PMID: 26335259 DOI: 10.1016/j.taap.2015.08.017] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2015] [Revised: 07/29/2015] [Accepted: 08/24/2015] [Indexed: 12/17/2022]
Abstract
The clinical efficacy of anthracycline anti-neoplastic agents is limited by cardiac and hepatic toxicities. The aim of this study was to assess the hepatoprotective and cardioprotective effects of taurine zinc solid dispersions, which is a newly-synthesized taurine zinc compound, against doxorubicin-induced toxicity in Sprague-Dawley rats intraperitoneally injected with doxorubicin hydrochloride (3mg/kg) three times a week (seven injections) over 28 days. Hemodynamic parameters, levels of liver toxicity markers and oxidative stress were assessed. Taurine zinc significantly attenuated the reductions in blood pressure, left ventricular pressure and ± dp/dtmax, increases in serum alanine aminotransferase and aspartate aminotransferase activities, and reductions in serum Zn(2+) and albumin levels (P<0.05 or 0.01) induced by doxorubicin. In rats treated with doxorubicin, taurine zinc dose-dependently increased liver superoxide dismutase activity and glutathione concentration, and decreased malondialdehyde level (P<0.01). qBase(+) was used to evaluate the stability of eight candidate reference genes for real-time quantitative reverse-transcription PCR. Taurine zinc dose-dependently increased liver heme oxygenase-1 and UDP-glucuronyl transferase mRNA and protein expression (P<0.01). Western blotting demonstrated that taurine zinc inhibited c-Jun N-terminal kinase phosphorylation by upregulating dual-specificity phosphoprotein phosphatase-1. Additionally, taurine zinc inhibited cardiomyocyte apoptosis as there was decreased TUNEL/DAPI positivity and protein expression of caspase-3. These results indicate that taurine zinc solid dispersions prevent the side-effects of anthracycline-based anticancer therapy. The mechanisms might be associated with the enhancement of antioxidant defense system partly through activating transcription to synthesize endogenous phase II medicine enzymes and anti-apoptosis through inhibiting JNK phosphorylation.
Collapse
Affiliation(s)
- Yu Wang
- Lab of Traditional Chinese Medicine and Marine Drugs, Department of Biochemistry, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, Guangdong, China
| | - Xueting Mei
- Lab of Traditional Chinese Medicine and Marine Drugs, Department of Biochemistry, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, Guangdong, China
| | - Jingquan Yuan
- Lab of Traditional Chinese Medicine and Marine Drugs, Department of Biochemistry, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, Guangdong, China
| | - Wenping Lu
- Lab of Traditional Chinese Medicine and Marine Drugs, Department of Biochemistry, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, Guangdong, China
| | - Binglong Li
- Lab of Traditional Chinese Medicine and Marine Drugs, Department of Biochemistry, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, Guangdong, China
| | - Donghui Xu
- Lab of Traditional Chinese Medicine and Marine Drugs, Department of Biochemistry, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, Guangdong, China.
| |
Collapse
|
203
|
Abstract
Cardiovascular complications are among the leading causes of morbidity and mortality among survivors of childhood cancer, after cancer relapse and secondary malignancies. Although advances in cancer treatment have improved the 5-year survival rates, the same treatments, such as anthracyclines, that cure cancer also increase the risk for adverse cardiovascular effects. Anthracycline-related cardiotoxicity in survivors of childhood cancer is progressive and can take years to develop, initially presenting as sub-clinical cardiac abnormalities that, if left undetected or untreated, can lead to heart failure, myocardial infarction, or other clinical cardiac dysfunction. A higher cumulative dose of anthracycline is associated with cardiotoxicity in children; however, sub-clinical cardiac abnormalities are evident at lower doses with longer follow-up, suggesting that there is no "safe" dose of anthracycline. Other risk factors include female sex, younger age at diagnosis, black race, trisomy 21, longer time since treatment, and the presence of pre-existing cardiovascular disease and co-morbidities. Cardioprotective strategies during treatment are limited in children. Enalapril provides only temporary cardioprotection, whereas continuous anthracycline infusion extends none. On the other hand, dexrazoxane successfully prevents or reduces anthracycline-related cardiotoxicity in children with cancer, without increased risks for recurrence of primary or second malignancies or reductions in anti-tumour efficacy. With more childhood cancer survivors now reaching adulthood, it is vital to understand the adverse effects of cancer treatment on the cardiovascular system and their long-term consequences to identify and establish optimal prevention and management strategies that balance oncologic efficacy with long-term safety.
Collapse
|
204
|
Koczor CA, Jiao Z, Fields E, Russ R, Ludaway T, Lewis W. AZT-induced mitochondrial toxicity: an epigenetic paradigm for dysregulation of gene expression through mitochondrial oxidative stress. Physiol Genomics 2015. [PMID: 26199398 DOI: 10.1152/physiolgenomics.00045.2015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Mitochondrial dysfunction causes oxidative stress and cardiomyopathy. Oxidative stress also is a side effect of dideoxynucleoside antiretrovirals (NRTI) and is observed in NRTI-induced cardiomyopathy. We show here that treatment with the NRTI AZT {1-[(2R,4S,5S)-4-azido-5-(hydroxymethyl)oxolan-2-yl]-5-methylpyrimidine-2,4-dione} modulates cardiac gene expression epigenetically through production of mitochondrially derived reactive oxygen species. Transgenic mice with ubiquitous expression of mitochondrially targeted catalase (MCAT) and C57Bl/6 wild-type mice littermates (WT) were administered AZT (0.22 mg/day po, 35 days), and cardiac DNA and mRNA were isolated. In AZT-treated WT, 95 cardiac genes were differentially expressed compared with vehicle-treated WTs. When MCAT mice were treated with AZT, each of those 95 genes reverted toward the expression of vehicle-treated WTs. In AZT-treated WT hearts, Mthfr [5,10-methylenetetrahydrofolate reductase; a critical enzyme in synthesis of methionine cycle intermediates including S-adenosylmethionine (SAM)], was overexpressed. Steady-state abundance of SAM in cardiac extracts from AZT-treated MCAT mice increased 60% above that of vehicle-treated MCAT. No such change occurred in WT. AZT caused hypermethylation (47%) and hypomethylation (53%) of differentially methylated DNA regions in WT cardiac DNA. AZT-treated MCAT heart DNA exhibited greater hypermethylation (91%) and less hypomethylation (9%) compared with vehicle-treated MCAT controls. The gene encoding protein kinase C-α displayed multifocal epigenetic regulation caused by oxidative stress. Results show that mitochondrially derived oxidative stress in the heart hinders cardiac DNA methylation, alters steady-state abundance of SAM, alters cardiac gene expression, and promotes characteristic pathophysiological changes of cardiomyopathy. This mechanism for NRTI toxicity offers insight into long-term side effects from these commonly used antiviral agents.
Collapse
Affiliation(s)
| | - Zhe Jiao
- Department of Pathology, Emory University, Atlanta, Georgia
| | - Earl Fields
- Department of Pathology, Emory University, Atlanta, Georgia
| | - Rodney Russ
- Department of Pathology, Emory University, Atlanta, Georgia
| | - Tomika Ludaway
- Department of Pathology, Emory University, Atlanta, Georgia
| | - William Lewis
- Department of Pathology, Emory University, Atlanta, Georgia
| |
Collapse
|
205
|
Dexrazoxane protects breast cancer patients with diabetes from chemotherapy-induced cardiotoxicity. Am J Med Sci 2015; 349:406-12. [PMID: 25723884 DOI: 10.1097/maj.0000000000000432] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND To evaluate the cardioprotective effect of dexrazoxane (DEX) on chemotherapy in patients with breast cancer with concurrent type 2 diabetes mellitus (DM2). METHODS Eighty female patients with breast cancer with DM2 were randomly assigned to receive chemotherapy only or chemotherapy plus DEX. All patients received 80 mg/m epirubicin and 500 mg/m cyclophosphamide by intravenous infusion every 3 weeks for a total of 6 cycles. The group assigned to receive chemotherapy alone received placebo 30 minutes before epirubicin administration. The group assigned to receive chemotherapy plus DEX received 800 mg/m DEX 30 minutes before epirubicin administration. Cardiac function and hematology before and after 6 cycles of chemotherapy were analyzed. RESULTS There was no difference in baseline systole or diastole function between the 2 DM2 groups. Patients receiving chemotherapy alone experienced significantly greater reductions in Ea and significantly greater elevations in E/Ea and Tei index in comparison with patients receiving chemotherapy plus DEX. After chemotherapy, superoxide dismutase was significantly reduced, and serum malondialdehyde (MDA) was significantly increased in patients with DM2. Serum superoxide dismutase levels were comparable between the 2 groups before and after chemotherapy, MDA levels were comparable between the 2 groups before chemotherapy, whereas serum MDA was significantly higher after chemotherapy in the chemotherapy alone group in comparison with the group that received DEX. CONCULSIONS DEX protects against cardiotoxicity induced by chemotherapy in patients with breast cancer with concurrent DM2.
Collapse
|
206
|
Abstract
The purpose of the present study was to compare the activity of two different clinically available iron chelators on the development of acute liver injury after administration of the bacterial endotoxin (lipopolysaccharide [LPS]) in rats. Lipopolysaccharide was administered either alone or after pretreatment with dexrazoxane (DEX) or deferoxamine (DFO). Control groups received only saline or its combination with either chelator. After 8 h, untreated LPS rats developed liver injury, with signs of inflammation and oxidative stress. Lipopolysaccharide reduced plasma iron concentrations in association with increased production of hepcidin and the reduced liver expression of ferroportin. Administration of chelating agents to LPS animals showed distinct effects. Although both drugs were able to reduce liver iron content, together with corresponding changes in hepcidin and ferroportin expressions, only DFO showed a protective effect against liver injury despite relatively small liver concentrations. In sharp contrast, DEX failed to improve any hallmark of liver injury and even worsened the GSH/GSSG ratio, the indicator of oxidative stress in the tissue. High-performance liquid chromatography-mass spectrometry analysis showed marked liver accumulation of iron-chelating metabolite of DEX (ADR-925), whereas the parent compound was undetectable. Further downregulation of transporters involved in bile formation was observed after DFO in the LPS group as well as in healthy animals. Neither chelator imposed significant liver injury in healthy animals. In conclusion, we demonstrated marked differences in the modulation of endotoxemic liver impairment between two iron chelators, implicating that particular qualities of chelating agents may be of crucial importance.
Collapse
|
207
|
Preclinical and clinical evidence for the role of resveratrol in the treatment of cardiovascular diseases. Biochim Biophys Acta Mol Basis Dis 2015; 1852:1155-77. [DOI: 10.1016/j.bbadis.2014.10.016] [Citation(s) in RCA: 211] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 10/24/2014] [Accepted: 10/27/2014] [Indexed: 12/12/2022]
|
208
|
Cyclovirobuxine D Attenuates Doxorubicin-Induced Cardiomyopathy by Suppression of Oxidative Damage and Mitochondrial Biogenesis Impairment. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:151972. [PMID: 26075032 PMCID: PMC4446494 DOI: 10.1155/2015/151972] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 02/11/2015] [Accepted: 02/23/2015] [Indexed: 01/04/2023]
Abstract
The clinical application of doxorubicin (DOX) is compromised by its cardiac toxic effect. Cyclovirobuxine D (CVB-D) is a steroid alkaloid extracted from a traditional Chinese
medicine, Buxus microphylla. Our results showed that CVB-D pretreatment markedly attenuated DOX-induced cardiac contractile dysfunction and histological alterations. By using TUNEL assay and western blot analysis, we found that CVB-D pretreatment reduced DOX-induced apoptosis of myocardial cells and
mitochondrial cytochrome c release to cytosol. CVB-D pretreatment ameliorated DOX-induced cardiac oxidative damage including lipid peroxidation and protein carbonylation and a decrease in the ratio of reduced glutathione (GSH) to oxidized glutathione (GSSG). Moreover, CVB-D was found to prevent DOX-induced mitochondrial biogenesis impairment as evidenced by preservation of peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α) and nuclear respiratory factor 1 (NRF1), as well as mitochondrial DNA copy number. These findings demonstrate that CVB-D protects against DOX-induced cardiomyopathy, at least in part, by suppression of oxidative damage and mitochondrial biogenesis impairment.
Collapse
|
209
|
Hao G, Yu Y, Gu B, Xing Y, Xue M. Protective effects of berberine against doxorubicin-induced cardiotoxicity in rats by inhibiting metabolism of doxorubicin. Xenobiotica 2015; 45:1024-9. [DOI: 10.3109/00498254.2015.1034223] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
210
|
Chen Y, Tang Y, Zhang YC, Huang XH, Xie YQ, Xiang Y. A metabolomic study of rats with doxorubicin-induced cardiomyopathy and Shengmai injection treatment. PLoS One 2015; 10:e0125209. [PMID: 25938766 PMCID: PMC4418690 DOI: 10.1371/journal.pone.0125209] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 03/23/2015] [Indexed: 01/06/2023] Open
Abstract
Doxorubicin-induced cardiomyopathy (DOX-CM) is a severe complication of doxorubicin (DOX) chemotherapy. Characterized by cumulative and irreversible myocardial damage, its pathogenesis has not been fully elucidated. Shengmai Injection (SMI), a Traditional Chinese Medicine, may alleviate myocardial injury and improve heart function in the setting of DOX-CM. As a result of its multi-component and multi-target nature and comprehensive regulation, the pharmacological mechanisms underlying SMI’s effects remain obscure. The emerging field of metabolomics provides a potential approach with which to explore the pathogenesis of DOX-CM and the benefits of SMI treatment. DOX-CM was induced in rats via intraperitoneal injections of DOX. Cardiac metabolic profiling was performed via gas chromatography/mass spectrometry and ultra-performance liquid chromatography/tandem mass spectrometry. A bioinformatics analysis was conducted via Ingenuity Pathway Analysis (IPA). Eight weeks following DOX treatment, significant cardiac remodeling, dysfunction and metabolic perturbations were observed in the rats with DOX-CM. The metabolic disturbances primarily involved lipids, amino acids, vitamins and energy metabolism, and may have been indicative of both an energy metabolism disorder and oxidative stress secondary to DOX chemotherapy. However, SMI improved cardiac structure and function, as well as the metabolism of the rats with DOX-CM. The metabolic alterations induced via SMI, including the promotion of glycogenolysis, glycolysis, amino acid utilization and antioxidation, suggested that SMI exerts cardioprotective effects by improving energy metabolism and attenuating oxidative stress. Moreover, the IPA revealed that important signaling molecules and enzymes interacted with the altered metabolites. These findings have provided us with new insights into the pathogenesis of DOX-CM and the effects of SMI, and suggest that the combination of metabolomic analysis and IPA may represent a promising tool with which to explore and better understand both heart disease and TCM therapy.
Collapse
Affiliation(s)
- Yu Chen
- The Division of Cardiology, Xin Hua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yong Tang
- The Division of Cardiology, Xin Hua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Ya-Chen Zhang
- The Division of Cardiology, Xin Hua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- * E-mail:
| | - Xiao-Hong Huang
- The Division of Cardiology, Xin Hua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yu-Quan Xie
- The Division of Cardiology, Xin Hua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yin Xiang
- The Division of Cardiology, Xin Hua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| |
Collapse
|
211
|
Rochette L, Guenancia C, Gudjoncik A, Hachet O, Zeller M, Cottin Y, Vergely C. Anthracyclines/trastuzumab: new aspects of cardiotoxicity and molecular mechanisms. Trends Pharmacol Sci 2015; 36:326-48. [PMID: 25895646 DOI: 10.1016/j.tips.2015.03.005] [Citation(s) in RCA: 172] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 03/16/2015] [Accepted: 03/20/2015] [Indexed: 01/26/2023]
Abstract
Anticancer drugs continue to cause significant reductions in left ventricular ejection fraction resulting in congestive heart failure. The best-known cardiotoxic agents are anthracyclines (ANTHs) such as doxorubicin (DOX). For several decades cardiotoxicity was almost exclusively associated with ANTHs, for which cumulative dose-related cardiac damage was the use-limiting step. Human epidermal growth factor (EGF) receptor 2 (HER2; ErbB2) has been identified as an important target for breast cancer. Trastuzumab (TRZ), a humanized anti-HER2 monoclonal antibody, is currently recommended as first-line treatment for patients with metastatic HER2(+) tumors. The use of TRZ may be limited by the development of drug intolerance, such as cardiac dysfunction. Cardiotoxicity has been attributed to free-iron-based, radical-induced oxidative stress. Many approaches have been promoted to minimize these serious side effects, but they are still clinically problematic. A new approach to personalized medicine for cancer that involves molecular screening for clinically relevant genomic alterations and genotype-targeted treatments is emerging.
Collapse
Affiliation(s)
- Luc Rochette
- Laboratoire de Physiopathologie et Pharmacologie Cardio-métaboliques (LPPCM), Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche 866, Facultés de Médecine et de Pharmacie - Université de Bourgogne, 7 Boulevard Jeanne d'Arc, 21033 Dijon, France.
| | - Charles Guenancia
- Laboratoire de Physiopathologie et Pharmacologie Cardio-métaboliques (LPPCM), Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche 866, Facultés de Médecine et de Pharmacie - Université de Bourgogne, 7 Boulevard Jeanne d'Arc, 21033 Dijon, France; Service de Cardiologie, Centre Hospitalier Universitaire Bocage, Dijon, France
| | - Aurélie Gudjoncik
- Laboratoire de Physiopathologie et Pharmacologie Cardio-métaboliques (LPPCM), Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche 866, Facultés de Médecine et de Pharmacie - Université de Bourgogne, 7 Boulevard Jeanne d'Arc, 21033 Dijon, France; Service de Cardiologie, Centre Hospitalier Universitaire Bocage, Dijon, France
| | - Olivier Hachet
- Laboratoire de Physiopathologie et Pharmacologie Cardio-métaboliques (LPPCM), Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche 866, Facultés de Médecine et de Pharmacie - Université de Bourgogne, 7 Boulevard Jeanne d'Arc, 21033 Dijon, France; Service de Cardiologie, Centre Hospitalier Universitaire Bocage, Dijon, France
| | - Marianne Zeller
- Laboratoire de Physiopathologie et Pharmacologie Cardio-métaboliques (LPPCM), Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche 866, Facultés de Médecine et de Pharmacie - Université de Bourgogne, 7 Boulevard Jeanne d'Arc, 21033 Dijon, France
| | - Yves Cottin
- Laboratoire de Physiopathologie et Pharmacologie Cardio-métaboliques (LPPCM), Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche 866, Facultés de Médecine et de Pharmacie - Université de Bourgogne, 7 Boulevard Jeanne d'Arc, 21033 Dijon, France; Service de Cardiologie, Centre Hospitalier Universitaire Bocage, Dijon, France
| | - Catherine Vergely
- Laboratoire de Physiopathologie et Pharmacologie Cardio-métaboliques (LPPCM), Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche 866, Facultés de Médecine et de Pharmacie - Université de Bourgogne, 7 Boulevard Jeanne d'Arc, 21033 Dijon, France
| |
Collapse
|
212
|
Moylan JS. Preserving muscle health and wellbeing for long-term cancer survivors. J Physiol 2015; 593:1767-8. [PMID: 25871558 DOI: 10.1113/jp270278] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Affiliation(s)
- Jennifer S Moylan
- Department of Physiology, Center for Muscle Biology, Center for Clinical and Translational Science, University of Kentucky, Lexington, KY, 40536, USA.
| |
Collapse
|
213
|
Iron-induced damage in cardiomyopathy: oxidative-dependent and independent mechanisms. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:230182. [PMID: 25878762 PMCID: PMC4387903 DOI: 10.1155/2015/230182] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Revised: 03/06/2015] [Accepted: 03/15/2015] [Indexed: 02/08/2023]
Abstract
The high incidence of cardiomyopathy in patients with hemosiderosis, particularly in transfusional iron overload, strongly indicates that iron accumulation in the heart plays a major role in the process leading to heart failure. In this context, iron-mediated generation of noxious reactive oxygen species is believed to be the most important pathogenetic mechanism determining cardiomyocyte damage, the initiating event of a pathologic progression involving apoptosis, fibrosis, and ultimately cardiac dysfunction. However, recent findings suggest that additional mechanisms involving subcellular organelles and inflammatory mediators are important factors in the development of this disease. Moreover, excess iron can amplify the cardiotoxic effect of other agents or events. Finally, subcellular misdistribution of iron within cardiomyocytes may represent an additional pathway leading to cardiac injury. Recent advances in imaging techniques and chelators development remarkably improved cardiac iron overload detection and treatment, respectively. However, increased understanding of the pathogenic mechanisms of iron overload cardiomyopathy is needed to pave the way for the development of improved therapeutic strategies.
Collapse
|
214
|
Cross MJ, Berridge BR, Clements PJM, Cove-Smith L, Force TL, Hoffmann P, Holbrook M, Lyon AR, Mellor HR, Norris AA, Pirmohamed M, Tugwood JD, Sidaway JE, Park BK. Physiological, pharmacological and toxicological considerations of drug-induced structural cardiac injury. Br J Pharmacol 2015; 172:957-74. [PMID: 25302413 PMCID: PMC4314188 DOI: 10.1111/bph.12979] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 09/30/2014] [Accepted: 10/06/2014] [Indexed: 01/01/2023] Open
Abstract
The incidence of drug-induced structural cardiotoxicity, which may lead to heart failure, has been recognized in association with the use of anthracycline anti-cancer drugs for many years, but has also been shown to occur following treatment with the new generation of targeted anti-cancer agents that inhibit one or more receptor or non-receptor tyrosine kinases, serine/threonine kinases as well as several classes of non-oncology agents. A workshop organized by the Medical Research Council Centre for Drug Safety Science (University of Liverpool) on 5 September 2013 and attended by industry, academia and regulatory representatives, was designed to gain a better understanding of the gaps in the field of structural cardiotoxicity that can be addressed through collaborative efforts. Specific recommendations from the workshop for future collaborative activities included: greater efforts to identify predictive (i) preclinical; and (ii) clinical biomarkers of early cardiovascular injury; (iii) improved understanding of comparative physiology/pathophysiology and the clinical predictivity of current preclinical in vivo models; (iv) the identification and use of a set of cardiotoxic reference compounds for comparative profiling in improved animal and human cellular models; (v) more sharing of data (through publication/consortia arrangements) on target-related toxicities; (vi) strategies to develop cardio-protective agents; and (vii) closer interactions between preclinical scientists and clinicians to help ensure best translational efforts.
Collapse
Affiliation(s)
- M J Cross
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of LiverpoolLiverpool, UK
| | - B R Berridge
- Safety Assessment, GlaxoSmithKlineResearch Triangle Park, NC, USA
| | - P J M Clements
- David Jack Centre for Research & Development, GlaxoSmithKlineWare, Herts, UK
| | - L Cove-Smith
- Clinical & Experimental Pharmacology, Cancer Research UK Manchester Institute, University of ManchesterManchester, UK
| | - T L Force
- Center for Translational Medicine and Cardiology Division, Temple University School of MedicinePhiladelphia, PA, USA
| | - P Hoffmann
- Preclinical Safety, Novartis Pharm CorpEast Hanover, NJ, USA
| | - M Holbrook
- Safety Pharmacology, Covance Laboratories, Ltd.Harrogate, North Yorkshire, UK
| | - A R Lyon
- NIHR Cardiovascular Biomedical Research Unit, Royal Brompton Hospital and Imperial CollegeLondon, UK
| | - H R Mellor
- Drug Safety Evaluation, Vertex Pharmaceuticals (Europe), Ltd.Abingdon, Oxfordshire, UK
| | - A A Norris
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of LiverpoolLiverpool, UK
| | - M Pirmohamed
- The Wolfson Centre for Personalised Medicine, Institute of Translational Medicine, University of LiverpoolLiverpool, UK
| | - J D Tugwood
- Clinical & Experimental Pharmacology, Cancer Research UK Manchester Institute, University of ManchesterManchester, UK
| | - J E Sidaway
- Innovative Medicines, AstraZeneca R&DMacclesfield, UK
| | - B K Park
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of LiverpoolLiverpool, UK
| |
Collapse
|
215
|
Zhou F, Hao G, Zhang J, Zheng Y, Wu X, Hao K, Niu F, Luo D, Sun Y, Wu L, Ye W, Wang G. Protective effect of 23-hydroxybetulinic acid on doxorubicin-induced cardiotoxicity: a correlation with the inhibition of carbonyl reductase-mediated metabolism. Br J Pharmacol 2015; 172:5690-703. [PMID: 25363561 DOI: 10.1111/bph.12995] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 10/17/2014] [Accepted: 10/27/2014] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND AND PURPOSE The clinical use of doxorubicin, an effective anticancer drug, is severely hampered by its cardiotoxicity. 23-Hydroxybetulinic acid (23-HBA), isolated from Pulsatilla chinensis, enhances the anticancer effect of doxorubicin while simultaneously reducing its cardiac toxicity, but does not affect the concentration of doxorubicin in the plasma and heart. As the metabolite doxorubicinol is more potent than doxorubicin at inducing cardiac toxicity, in the present study we aimed to clarify the role of doxorubicinol in the protective effect of 23-HBA. EXPERIMENTAL APPROACH Doxorubicin was administered to mice for two weeks in the presence or absence of 23-HBA. The heart pathology, function, myocardial enzymes and accumulation of doxorubicin and doxorubicinol were then analysed. A cellular pharmacokinetic study of doxorubicin and doxorubicinol, carbonyl reductase 1 (CBR1) interference and molecular docking was performed in vitro. KEY RESULTS 23-HBA alleviated the doxorubicin-induced cardiotoxicity in mice, and this was accompanied by inhibition of the metabolism of doxorubicin and reduced accumulation of doxorubicinol selectively in hearts. In H9c2 cells, the protective effect of 23-HBA was shown to be closely associated with a decreased rate and extent of accumulation of doxorubicinol in mitochondria and nuclei. siRNA and docking analysis demonstrated that CBR1 has a crucial role in doxorubicin-mediated cardiotoxicity and 23-HBA inhibits this metabolic pathway. CONCLUSIONS AND IMPLICATIONS Inhibition of CBR-mediated doxorubicin metabolism might be one of the protective mechanisms of 23-HBA against doxorubicin-induced cardiotoxicity. The present study provides a new research strategy guided by pharmacokinetic theory to elucidate the mechanism of drugs with unknown targets.
Collapse
Affiliation(s)
- Fang Zhou
- Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China
| | - Gang Hao
- Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China.,Suzhou Institute for Food and Drug Control, Suzhou, China
| | - Jingwei Zhang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China
| | - Yuanting Zheng
- Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China
| | - Xiaolan Wu
- Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China
| | - Kun Hao
- Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China
| | - Fang Niu
- Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China
| | - Dan Luo
- Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China
| | - Yuan Sun
- Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China
| | - Liang Wu
- Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China
| | - Wencai Ye
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Guangji Wang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China.,Jiangsu Key laboratory of drug design and optimization, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
216
|
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 DOI: 10.2119/molmed.2014.00261] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [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
|
217
|
Moulin M, Piquereau J, Mateo P, Fortin D, Rucker-Martin C, Gressette M, Lefebvre F, Gresikova M, Solgadi A, Veksler V, Garnier A, Ventura-Clapier R. Sexual Dimorphism of Doxorubicin-Mediated Cardiotoxicity. Circ Heart Fail 2015; 8:98-108. [DOI: 10.1161/circheartfailure.114.001180] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Background—
Cardiovascular diseases are the major cause of mortality among both men and women with a lower incidence in women before menopause. The clinical use of doxorubicin, widely used as an antineoplastic agent, is markedly hampered by severe cardiotoxicity. Even if there is a significant sex difference in incidence of cardiovascular disease at the adult stage, it is not known whether a difference in doxorubicin-related cardiotoxicity between men and women also exists. The objective of this work was to explore the cardiac side effects of doxorubicin in adult rats and decipher whether signaling pathways involved in cardiac toxicity differ between sexes.
Methods and Results—
After 7 weeks of doxorubicin (2 mg/kg per week), males developed major signs of cardiomyopathy with cardiac atrophy, reduced left ventricular ejection fraction and 50% mortality. In contrast, no female died and their left ventricular ejection fraction was only moderately affected. Surprisingly, neither global oxidation levels nor the antioxidant response nor the apoptosis signaling pathways were altered by doxorubicin. However, the level of total adenosine monophosphate–activated protein kinase was severely decreased only in males. Moreover, markers of mitochondrial biogenesis and cardiolipin content were strongly reduced only in males. To analyze the onset of the pathology, maximal oxygen consumption rate of left ventricular permeabilized fibers after 4 weeks of treatment was reduced only in doxorubicin-treated males.
Conclusions—
Altogether, these results clearly evidence sex differences in doxorubicin toxicity. Cardiac mitochondrial dysfunction and adenosine monophosphate–activated protein kinase seem as critical sites of sex differences in cardiotoxicity as evidenced by significant statistical interactions between sex and treatment effects.
Collapse
Affiliation(s)
- Maryline Moulin
- From the INSERM UMR-S 769, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., M.G., F.L., M.G., V.V., A.G., R.V.-C.); IPSIT-IFR141 Université de Paris-Sud, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., C.R.-M., M.G., F.L., M.G., A.S., V.V., A.G., R.V.-C.); INSERM UMR-S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France (C.R.-M.); and IPSIT-IFR141 Service d’Analyse des Médicaments et Métabolites, Châtenay-Malabry, France (A.S.)
| | - Jérôme Piquereau
- From the INSERM UMR-S 769, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., M.G., F.L., M.G., V.V., A.G., R.V.-C.); IPSIT-IFR141 Université de Paris-Sud, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., C.R.-M., M.G., F.L., M.G., A.S., V.V., A.G., R.V.-C.); INSERM UMR-S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France (C.R.-M.); and IPSIT-IFR141 Service d’Analyse des Médicaments et Métabolites, Châtenay-Malabry, France (A.S.)
| | - Philippe Mateo
- From the INSERM UMR-S 769, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., M.G., F.L., M.G., V.V., A.G., R.V.-C.); IPSIT-IFR141 Université de Paris-Sud, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., C.R.-M., M.G., F.L., M.G., A.S., V.V., A.G., R.V.-C.); INSERM UMR-S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France (C.R.-M.); and IPSIT-IFR141 Service d’Analyse des Médicaments et Métabolites, Châtenay-Malabry, France (A.S.)
| | - Dominique Fortin
- From the INSERM UMR-S 769, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., M.G., F.L., M.G., V.V., A.G., R.V.-C.); IPSIT-IFR141 Université de Paris-Sud, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., C.R.-M., M.G., F.L., M.G., A.S., V.V., A.G., R.V.-C.); INSERM UMR-S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France (C.R.-M.); and IPSIT-IFR141 Service d’Analyse des Médicaments et Métabolites, Châtenay-Malabry, France (A.S.)
| | - Catherine Rucker-Martin
- From the INSERM UMR-S 769, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., M.G., F.L., M.G., V.V., A.G., R.V.-C.); IPSIT-IFR141 Université de Paris-Sud, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., C.R.-M., M.G., F.L., M.G., A.S., V.V., A.G., R.V.-C.); INSERM UMR-S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France (C.R.-M.); and IPSIT-IFR141 Service d’Analyse des Médicaments et Métabolites, Châtenay-Malabry, France (A.S.)
| | - Mélanie Gressette
- From the INSERM UMR-S 769, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., M.G., F.L., M.G., V.V., A.G., R.V.-C.); IPSIT-IFR141 Université de Paris-Sud, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., C.R.-M., M.G., F.L., M.G., A.S., V.V., A.G., R.V.-C.); INSERM UMR-S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France (C.R.-M.); and IPSIT-IFR141 Service d’Analyse des Médicaments et Métabolites, Châtenay-Malabry, France (A.S.)
| | - Florence Lefebvre
- From the INSERM UMR-S 769, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., M.G., F.L., M.G., V.V., A.G., R.V.-C.); IPSIT-IFR141 Université de Paris-Sud, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., C.R.-M., M.G., F.L., M.G., A.S., V.V., A.G., R.V.-C.); INSERM UMR-S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France (C.R.-M.); and IPSIT-IFR141 Service d’Analyse des Médicaments et Métabolites, Châtenay-Malabry, France (A.S.)
| | - Milada Gresikova
- From the INSERM UMR-S 769, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., M.G., F.L., M.G., V.V., A.G., R.V.-C.); IPSIT-IFR141 Université de Paris-Sud, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., C.R.-M., M.G., F.L., M.G., A.S., V.V., A.G., R.V.-C.); INSERM UMR-S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France (C.R.-M.); and IPSIT-IFR141 Service d’Analyse des Médicaments et Métabolites, Châtenay-Malabry, France (A.S.)
| | - Audrey Solgadi
- From the INSERM UMR-S 769, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., M.G., F.L., M.G., V.V., A.G., R.V.-C.); IPSIT-IFR141 Université de Paris-Sud, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., C.R.-M., M.G., F.L., M.G., A.S., V.V., A.G., R.V.-C.); INSERM UMR-S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France (C.R.-M.); and IPSIT-IFR141 Service d’Analyse des Médicaments et Métabolites, Châtenay-Malabry, France (A.S.)
| | - Vladimir Veksler
- From the INSERM UMR-S 769, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., M.G., F.L., M.G., V.V., A.G., R.V.-C.); IPSIT-IFR141 Université de Paris-Sud, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., C.R.-M., M.G., F.L., M.G., A.S., V.V., A.G., R.V.-C.); INSERM UMR-S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France (C.R.-M.); and IPSIT-IFR141 Service d’Analyse des Médicaments et Métabolites, Châtenay-Malabry, France (A.S.)
| | - Anne Garnier
- From the INSERM UMR-S 769, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., M.G., F.L., M.G., V.V., A.G., R.V.-C.); IPSIT-IFR141 Université de Paris-Sud, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., C.R.-M., M.G., F.L., M.G., A.S., V.V., A.G., R.V.-C.); INSERM UMR-S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France (C.R.-M.); and IPSIT-IFR141 Service d’Analyse des Médicaments et Métabolites, Châtenay-Malabry, France (A.S.)
| | - Renée Ventura-Clapier
- From the INSERM UMR-S 769, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., M.G., F.L., M.G., V.V., A.G., R.V.-C.); IPSIT-IFR141 Université de Paris-Sud, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., C.R.-M., M.G., F.L., M.G., A.S., V.V., A.G., R.V.-C.); INSERM UMR-S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France (C.R.-M.); and IPSIT-IFR141 Service d’Analyse des Médicaments et Métabolites, Châtenay-Malabry, France (A.S.)
| |
Collapse
|
218
|
Jirkovská-Vávrová A, Roh J, Lenčová-Popelová O, Jirkovský E, Hrušková K, Potůčková-Macková E, Jansová H, Hašková P, Martinková P, Eisner T, Kratochvíl M, Šůs J, Macháček M, Vostatková-Tichotová L, Geršl V, Kalinowski DS, Muller MT, Richardson DR, Vávrová K, Štěrba M, Šimůnek T. Synthesis and analysis of novel analogues of dexrazoxane and its open-ring hydrolysis product for protection against anthracycline cardiotoxicity in vitro and in vivo. Toxicol Res (Camb) 2015. [DOI: 10.1039/c5tx00048c] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Topoisomerase II beta, rather than (or along with) iron chelation, may be a promising target for cardioprotection.
Collapse
|
219
|
Bureš J, Jansová H, Stariat J, Filipský T, Mladěnka P, Šimůnek T, Kučera R, Klimeš J, Wang Q, Franz KJ, Kovaříková P. LC-UV/MS methods for the analysis of prochelator-boronyl salicylaldehyde isonicotinoyl hydrazone (BSIH) and its active chelator salicylaldehyde isonicotinoyl hydrazone (SIH). J Pharm Biomed Anal 2014; 105:55-63. [PMID: 25527982 DOI: 10.1016/j.jpba.2014.11.044] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 11/20/2014] [Accepted: 11/25/2014] [Indexed: 01/06/2023]
Abstract
Salicylaldehyde isonicotinoyl hydrazone (SIH) is an intracellular iron chelator with well documented potential to protect against oxidative injury both in vitro and in vivo. However, it suffers from short biological half-life caused by fast hydrolysis of the hydrazone bond. Recently, a concept of boronate prochelators has been introduced as a strategy that might overcome these limitations. This study presents two complementary analytical methods for detecting the prochelator-boronyl salicylaldehyde isonicotinoyl hydrazone-BSIH along with its active metal-binding chelator SIH in different solution matrices and concentration ranges. An LC-UV method for determination of BSIH and SIH in buffer and cell culture medium was validated over concentrations of 7-115 and 4-115 μM, respectively, and applied to BSIH activation experiments in vitro. An LC-MS assay was validated for quantification of BSIH and SIH in plasma over the concentration range of 0.06-23 and 0.24-23 μM, respectively, and applied to stability studies in plasma in vitro as well as analysis of plasma taken after i.v. administration of BSIH to rats. A Zorbax-RP bonus column and mobile phases containing either phosphate buffer with EDTA or ammonium formate and methanol/acetonitrile mixture provided suitable conditions for the LC-UV and LC-MS analysis, respectively. Samples were diluted or precipitated with methanol prior to analysis. These separative analytical techniques establish the first validated protocols to investigate BSIH activation by hydrogen peroxide in multiple matrices, directly compare the stabilities of the prochelator and its chelator in plasma, and provide the first basic pharmacokinetic data of this prochelator. Experiments reveal that BSIH is stable in all media tested and is partially converted to SIH by H2O2. The observed integrity of BSIH in plasma samples from the in vivo study suggests that the concept of prochelation might be a promising strategy for further development of aroylhydrazone cytoprotective agents.
Collapse
Affiliation(s)
- Jan Bureš
- Faculty of Pharmacy in Hradec Králové, Charles University in Prague, Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| | - Hana Jansová
- Faculty of Pharmacy in Hradec Králové, Charles University in Prague, Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| | - Ján Stariat
- Faculty of Pharmacy in Hradec Králové, Charles University in Prague, Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| | - Tomáš Filipský
- Faculty of Pharmacy in Hradec Králové, Charles University in Prague, Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| | - Přemysl Mladěnka
- Faculty of Pharmacy in Hradec Králové, Charles University in Prague, Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| | - Tomáš Šimůnek
- Faculty of Pharmacy in Hradec Králové, Charles University in Prague, Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| | - Radim Kučera
- Faculty of Pharmacy in Hradec Králové, Charles University in Prague, Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| | - Jiří Klimeš
- Faculty of Pharmacy in Hradec Králové, Charles University in Prague, Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| | - Qin Wang
- Duke University, Department of Chemistry, Durham, NC 22708, USA
| | | | - Petra Kovaříková
- Faculty of Pharmacy in Hradec Králové, Charles University in Prague, Heyrovského 1203, 500 05 Hradec Králové, Czech Republic.
| |
Collapse
|
220
|
Sodium ferulate protects against daunorubicin-induced cardiotoxicity by inhibition of mitochondrial apoptosis in juvenile rats. J Cardiovasc Pharmacol 2014; 63:360-8. [PMID: 24336018 DOI: 10.1097/fjc.0000000000000056] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Daunorubicin (DNR) is a widely used chemotherapeutic agent; however, its clinical use is limited because of its cardiotoxicity. This study was aimed to investigate the protective effect of sodium ferulate (SF), an effective component from traditional Chinese herbs, against DNR-induced cardiotoxicity in juvenile rats. DNR was administered intraperitoneally to rats at the dosage of 2.5 mg·kg(-1)·wk(-1) for 5 consecutive weeks (cumulative dose of 12.5 mg/kg) or in combination with intraperitoneal injection of SF at 50 mg·kg(-1)·d(-1) over a period of 30 days. The animals were killed 6 days after the last injection of DNR. SF significantly ameliorated the DNR-induced cardiac dysfunction, structural damage of the myocardium, and release of lactate dehydrogenase and creatine kinase. Treatment with SF also reversed DNR-induced oxidative stress as evidenced by a decrease in malondialdehyde levels with a concomitant increase in myocardical superoxide dismutase activities. Furthermore, SF afforded significant cardioprotection against DNR-induced apoptosis in vivo and effectively suppressed the complex mitochondrion-dependent apoptotic signaling triggered by DNR. This study indicates that SF may improve cardiac function by inhibition of oxidative stress and apoptosis, thus providing a beneficial effect on the prevention of DNR-induced cardiotoxicity.
Collapse
|
221
|
Hsu HC, Chen CY, Chen MF. N-3 polyunsaturated fatty acids decrease levels of doxorubicin-induced reactive oxygen species in cardiomyocytes -- involvement of uncoupling protein UCP2. J Biomed Sci 2014; 21:101. [PMID: 25407516 PMCID: PMC4237738 DOI: 10.1186/s12929-014-0101-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 10/31/2014] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Use of the chemotherapeutic drug doxorubicin (DOX) is associated with serious cardiotoxicity, as it increases levels of reactive oxygen species (ROS). N-3 polyunsaturated fatty acid dietary supplements can be of benefit to patients undergoing cancer therapy. The aims of this study were to determine whether DOX-induced cardiotoxicity is related to mitochondrial uncoupling proteins and whether eicosapentaenoic acid (EPA, C20:5 n-3) or docosahexaenoic acid (DHA, C22:6 n-3) affects DOX-induced cardiomyocyte toxicity. RESULTS Treatment of H9C2 cells with DOX resulted in decreased cell viability and UCP2 expression. Treatment with 100 μM EPA or 50 μM DHA for 24 h resulted in a maximal mitochondria concentration of these fatty acids and increased UCP2 expression. Pretreatment with 100 μM EPA or 50 μM DHA prevented the DOX-induced decrease in UCP2 mRNA and protein levels, but these effects were not seen with EPA or DHA and DOX cotreatment. In addition, the DOX-induced increase in ROS production and subsequent mitochondrial membrane potential change (∆ψ) were significantly attenuated by pretreatment with EPA or DHA. CONCLUSION EPA or DHA pre-treatment inhibits the DOX-induced decrease in UCP2 expression, increase in ROS production, and subsequent mitochondrial membrane potential change that contribute to the cardiotoxicity of DOX.
Collapse
Affiliation(s)
- Hsiu-Ching Hsu
- />Department of Internal Medicine, National Taiwan University Hospital, 7 Chung-Shan S Rd, Taipei, Taiwan
| | - Ching-Yi Chen
- />Department of Animal Science and Technology, National Taiwan University, No. 50, Lane 155, Sec 3, Keelung Rd, Taipei, 10672 Taiwan
- />Present address: 50, Lane 155, Sec 3, Keelung Rd, Taipei, 106 Taiwan
| | - Ming-Fong Chen
- />Department of Internal Medicine, National Taiwan University Hospital, 7 Chung-Shan S Rd, Taipei, Taiwan
| |
Collapse
|
222
|
Gao S, Li H, Feng XJ, Li M, Liu ZP, Cai Y, Lu J, Huang XY, Wang JJ, Li Q, Chen SR, Ye JT, Liu PQ. α-Enolase plays a catalytically independent role in doxorubicin-induced cardiomyocyte apoptosis and mitochondrial dysfunction. J Mol Cell Cardiol 2014; 79:92-103. [PMID: 25446184 DOI: 10.1016/j.yjmcc.2014.11.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 10/29/2014] [Accepted: 11/05/2014] [Indexed: 12/30/2022]
Abstract
BACKGROUND α-Enolase is a glycolytic enzyme with "second jobs" beyond its catalytic activity. However, its possible contribution to cardiac dysfunction remains to be determined. The present study aimed to investigate the role of α-enolase in doxorubicin (Dox)-induced cardiomyopathy as well as the underlying mechanisms. EXPERIMENTAL APPROACHES The expression of α-enolase was detected in rat hearts and primary cultured rat cardiomyocytes with or without Dox administration. An adenovirus carrying short-hairpin interfering RNA targeting α-enolase was constructed and transduced specifically into the heart by intramyocardial injection. Heart function, cell apoptosis and mitochondrial function were measured following Dox administration. In addition, by using gain- and loss-of-function approaches to regulate α-enolase expression in primary cultured rat cardiomyocytes, we investigated the role of endogenous, wide type and catalytically inactive mutant α-enolase in cardiomyocyte apoptosis and ATP generation. Furthermore, the involvement of α-enolase in AMPK phosphorylation was also studied. KEY RESULTS The mRNA and protein expression of cardiac α-enolase was significantly upregulated by Dox. Genetic silencing of α-enolase in rat hearts and cultured cardiomyocytes attenuated Dox-induced apoptosis and mitochondrial dysfunction. In contrast, overexpression of wide-type or catalytically inactive α-enolase in cardiomyocytes mimicked the detrimental role of Dox in inducing apoptosis and ATP reduction. AMPK dephosphorylation was further demonstrated to be involved in the proapoptotic and ATP-depriving effects of α-enolase. CONCLUSION Our findings provided the evidence that α-enolase has a catalytically independent role in inducing cardiomyocyte apoptosis and mitochondrial dysfunction, which could be at least partially contributed to the inhibition of AMPK phosphorylation.
Collapse
Affiliation(s)
- Si Gao
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Higher Education Mega Center, No. 132 East Wai-huan Road, Guangzhou 510006, Guangdong, PR China; School of Medicine, Guangxi University of Science and Technology, No. 257 Liu-shi Road, Liuzhou 545005, Guangxi, PR China
| | - Hong Li
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Higher Education Mega Center, No. 132 East Wai-huan Road, Guangzhou 510006, Guangdong, PR China
| | - Xiao-jun Feng
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Higher Education Mega Center, No. 132 East Wai-huan Road, Guangzhou 510006, Guangdong, PR China
| | - Min Li
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Higher Education Mega Center, No. 132 East Wai-huan Road, Guangzhou 510006, Guangdong, PR China
| | - Zhi-ping Liu
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Higher Education Mega Center, No. 132 East Wai-huan Road, Guangzhou 510006, Guangdong, PR China
| | - Yi Cai
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Higher Education Mega Center, No. 132 East Wai-huan Road, Guangzhou 510006, Guangdong, PR China; Guangzhou Research Institute of Snake Venom, Guangzhou Medical College, Guangzhou 510182, Guangdong, PR China
| | - Jing Lu
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Higher Education Mega Center, No. 132 East Wai-huan Road, Guangzhou 510006, Guangdong, PR China
| | - Xiao-yang Huang
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Higher Education Mega Center, No. 132 East Wai-huan Road, Guangzhou 510006, Guangdong, PR China
| | - Jiao-jiao Wang
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Higher Education Mega Center, No. 132 East Wai-huan Road, Guangzhou 510006, Guangdong, PR China
| | - Qin Li
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Higher Education Mega Center, No. 132 East Wai-huan Road, Guangzhou 510006, Guangdong, PR China
| | - Shao-rui Chen
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Higher Education Mega Center, No. 132 East Wai-huan Road, Guangzhou 510006, Guangdong, PR China
| | - Jian-tao Ye
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Higher Education Mega Center, No. 132 East Wai-huan Road, Guangzhou 510006, Guangdong, PR China.
| | - Pei-qing Liu
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Higher Education Mega Center, No. 132 East Wai-huan Road, Guangzhou 510006, Guangdong, PR China.
| |
Collapse
|
223
|
Zhang YY, Meng C, Zhang XM, Yuan CH, Wen MD, Chen Z, Dong DC, Gao YH, Liu C, Zhang Z. Ophiopogonin D attenuates doxorubicin-induced autophagic cell death by relieving mitochondrial damage in vitro and in vivo. J Pharmacol Exp Ther 2014; 352:166-74. [PMID: 25378375 DOI: 10.1124/jpet.114.219261] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
It has been reported that ophiopogonin D (OP-D), a steroidal glycoside and an active component extracted from Ophiopogon japonicas, promotes antioxidative protection of the cardiovascular system. However, it is unknown whether OP-D exerts protective effects against doxorubicin (DOX)-induced autophagic cardiomyocyte injury. Here, we demonstrate that DOX induced excessive autophagy through the generation of reactive oxygen species (ROS) in H9c2 cells and in mouse hearts, which was indicated by a significant increase in the number of autophagic vacuoles, LC3-II/LC3-I ratio, and upregulation of the expression of GFP-LC3. Pretreatment with OP-D partially attenuated the above phenomena, similar to the effects of treatment with 3-methyladenine. In addition, OP-D treatment significantly relieved the disruption of the mitochondrial membrane potential by antioxidative effects through downregulating the expression of both phosphorylated c-Jun N-terminal kinase and extracellular signal-regulated kinase. The ability of OP-D to reduce the generation of ROS due to mitochondrial damage and, consequently, to inhibit autophagic activity partially accounts for its protective effects in the hearts against DOX-induced toxicity.
Collapse
Affiliation(s)
- Ying-Yu Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China (Y.-Y.Z., C.M., C.-H.Y., M.-D.W., Z.C., D.-C.D., Y.-H.G., C.L., Z.Z.); Department of Clinical Medicine, Changchun Medical College, Changchun, China (Y.-Y.Z.); and Department of Biopharmaceutical, School of Pharmacy, Jilin University, Changchun, China (X.-M.Z)
| | - Chen Meng
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China (Y.-Y.Z., C.M., C.-H.Y., M.-D.W., Z.C., D.-C.D., Y.-H.G., C.L., Z.Z.); Department of Clinical Medicine, Changchun Medical College, Changchun, China (Y.-Y.Z.); and Department of Biopharmaceutical, School of Pharmacy, Jilin University, Changchun, China (X.-M.Z)
| | - Xin-Mu Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China (Y.-Y.Z., C.M., C.-H.Y., M.-D.W., Z.C., D.-C.D., Y.-H.G., C.L., Z.Z.); Department of Clinical Medicine, Changchun Medical College, Changchun, China (Y.-Y.Z.); and Department of Biopharmaceutical, School of Pharmacy, Jilin University, Changchun, China (X.-M.Z)
| | - Cai-Hua Yuan
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China (Y.-Y.Z., C.M., C.-H.Y., M.-D.W., Z.C., D.-C.D., Y.-H.G., C.L., Z.Z.); Department of Clinical Medicine, Changchun Medical College, Changchun, China (Y.-Y.Z.); and Department of Biopharmaceutical, School of Pharmacy, Jilin University, Changchun, China (X.-M.Z)
| | - Ming-Da Wen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China (Y.-Y.Z., C.M., C.-H.Y., M.-D.W., Z.C., D.-C.D., Y.-H.G., C.L., Z.Z.); Department of Clinical Medicine, Changchun Medical College, Changchun, China (Y.-Y.Z.); and Department of Biopharmaceutical, School of Pharmacy, Jilin University, Changchun, China (X.-M.Z)
| | - Zhong Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China (Y.-Y.Z., C.M., C.-H.Y., M.-D.W., Z.C., D.-C.D., Y.-H.G., C.L., Z.Z.); Department of Clinical Medicine, Changchun Medical College, Changchun, China (Y.-Y.Z.); and Department of Biopharmaceutical, School of Pharmacy, Jilin University, Changchun, China (X.-M.Z)
| | - Da-Chuan Dong
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China (Y.-Y.Z., C.M., C.-H.Y., M.-D.W., Z.C., D.-C.D., Y.-H.G., C.L., Z.Z.); Department of Clinical Medicine, Changchun Medical College, Changchun, China (Y.-Y.Z.); and Department of Biopharmaceutical, School of Pharmacy, Jilin University, Changchun, China (X.-M.Z)
| | - Yan-Hong Gao
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China (Y.-Y.Z., C.M., C.-H.Y., M.-D.W., Z.C., D.-C.D., Y.-H.G., C.L., Z.Z.); Department of Clinical Medicine, Changchun Medical College, Changchun, China (Y.-Y.Z.); and Department of Biopharmaceutical, School of Pharmacy, Jilin University, Changchun, China (X.-M.Z)
| | - Chang Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China (Y.-Y.Z., C.M., C.-H.Y., M.-D.W., Z.C., D.-C.D., Y.-H.G., C.L., Z.Z.); Department of Clinical Medicine, Changchun Medical College, Changchun, China (Y.-Y.Z.); and Department of Biopharmaceutical, School of Pharmacy, Jilin University, Changchun, China (X.-M.Z)
| | - Zhao Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China (Y.-Y.Z., C.M., C.-H.Y., M.-D.W., Z.C., D.-C.D., Y.-H.G., C.L., Z.Z.); Department of Clinical Medicine, Changchun Medical College, Changchun, China (Y.-Y.Z.); and Department of Biopharmaceutical, School of Pharmacy, Jilin University, Changchun, China (X.-M.Z)
| |
Collapse
|
224
|
Czepas J, Gwoździński K. The flavonoid quercetin: possible solution for anthracycline-induced cardiotoxicity and multidrug resistance. Biomed Pharmacother 2014; 68:1149-59. [PMID: 25458790 DOI: 10.1016/j.biopha.2014.10.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 10/16/2014] [Indexed: 02/06/2023] Open
Abstract
Anthracycline chemotherapy is often used in the treatment of various malignancies. Its application, however, encounters several limitations due to development of serious side effects, mainly cardiotoxicity and may be ineffective due to multidrug resistance (MDR). Many different compounds have been evaluated as poorly effective in the protection against anthracycline side effects and in the prevention from MDR. Thus, continuous investigational efforts are necessary to find valuable protectants and the flavonoid quercetin (Q) seems to be a promising candidate. It is present in relatively high amounts in a human diet and the lack of its toxicity, including genotoxicity has been confirmed. The structure of Q favours its high antioxidant activity, the potential to inhibit the activity of oxidative enzymes and to interact with membrane transporter proteins responsible for development of MDR, e.g. P-glycoprotein. Furthermore, Q can influence cellular signalling and gene expression, and thus, alter response to exogenous genotoxicants and oxidative stress in normal cells. It accounts for its chemopreventive and anticancer properties. Overall, these properties might indicate the possibility of application of Q as cardioprotectant during anthracycline chemotherapy. Moreover, numerous biological properties displayed by Q might possibly result in the reversal of MDR in tumour cells and improve the efficacy of chemotherapy. However, these beneficial effects towards anthracycline-induced complications of chemotherapy have to be further explored and confirmed both in animal and clinical studies. Concurrently, investigations aimed at improvement of the bioavailability of Q and further elucidation of its metabolism after application in combination with anthracyclines are needed.
Collapse
Affiliation(s)
- Jan Czepas
- Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Łódź, Łódź, Poland.
| | - Krzysztof Gwoździński
- Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Łódź, Łódź, Poland
| |
Collapse
|
225
|
Szwed M, Kania KD, Jozwiak Z. Molecular damage caused by generation of reactive oxygen species in the redox cycle of doxorubicin-transferrin conjugate in human leukemia cell lines. Leuk Lymphoma 2014; 56:1475-83. [PMID: 25166006 DOI: 10.3109/10428194.2014.955022] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
In this study we focused on evaluation of the pro-oxidant properties of doxorubicin-transferrin (DOX-TRF) conjugate and its potency to damage macromolecules which are components of cellular compartments. Our experiments were performed on two human leukemia cell lines: K562 (chronic erythromyeloblastoid leukemia) and CCRF-CEM (acute lymphoblastic leukemia). We determined the reactive oxygen species (ROS) production and programmed cell death (PCD) induction by free DOX and its conjugate. Besides this, the lipid peroxidation and protein damage which can be provoked by DOX alone and DOX-TRF conjugate were assessed. ROS were produced in leukemia cells incubated with free DOX and DOX-TRF conjugate and the extent of apoptosis and necrosis was strongly dependent on the cell line, sensitivity to drug and time of incubation with the investigated compounds. The role of ROS in DOX-TRF conjugate-induced cell death was confirmed by the diminution effects of the antioxidant vitamin C.
Collapse
Affiliation(s)
- Marzena Szwed
- Department of Thermobiology, Faculty of Biology and Environmental Protection, University of Lodz , Lodz , Poland
| | | | | |
Collapse
|
226
|
Zamperone A, Pietronave S, Colangelo D, Antonini S, Locatelli M, Travaglia F, Coïsson JD, Arlorio M, Prat M. Protective effects of clovamide against H2O2-induced stress in rat cardiomyoblasts H9c2 cell line. Food Funct 2014; 5:2542-51. [PMID: 25133994 DOI: 10.1039/c4fo00195h] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Cocoa contains phenolic compounds with known antioxidant and antiradical properties beneficial in different pathologies, including cardiovascular diseases. Herein, we have evaluated the protective effects of clovamide, a minor cocoa component, against oxidative stress induced in the rat cardiomyoblast cell line, also comparing it to its bio-isosteric form, rosmarinic acid, and to the main monomeric flavan-3-ol from low-molecular-weight polyphenol in cocoa, i.e. epicatechin. At nano-micro-molar concentrations, the three compounds inhibited the production of reactive oxygen species and apoptosis, evaluated under different aspects, namely, annexin V positivity, DNA fragmentation, caspase release and activation. These molecules can, thus, be considered for their bioactive beneficial activity in the context of cardiovascular pathologies and, particularly, in the protection towards oxidative stress that follows ischemic injury. Clovamide may, thus, be the primary compound for the development of innovative nutraceutical strategies towards cardiovascular diseases.
Collapse
Affiliation(s)
- Andrea Zamperone
- Department of Health Sciences, Università del Piemonte Orientale "A. Avogadro", Novara, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
227
|
Polk A, Vistisen K, Vaage-Nilsen M, Nielsen DL. A systematic review of the pathophysiology of 5-fluorouracil-induced cardiotoxicity. BMC Pharmacol Toxicol 2014; 15:47. [PMID: 25186061 PMCID: PMC4170068 DOI: 10.1186/2050-6511-15-47] [Citation(s) in RCA: 160] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 08/19/2014] [Indexed: 12/29/2022] Open
Abstract
Background Cardiotoxicity is a serious side effect to treatment with 5-fluorouracil (5-FU), but the underlying mechanisms are not fully understood. The objective of this systematic review was to evaluate the pathophysiology of 5-FU- induced cardiotoxicity. Methods We systematically searched PubMed for articles in English using the search terms: 5-FU OR 5-fluorouracil OR capecitabine AND cardiotoxicity. Papers evaluating the pathophysiology of this cardiotoxicity were included. Results We identified 27 articles of 26 studies concerning the pathophysiology of 5-FU-induced cardiotoxicity. The studies demonstrated 5-FU-induced: hemorrhagic infarction, interstitial fibrosis and inflammatory reaction in the myocardium; damage of the arterial endothelium followed by platelet aggregation; increased myocardial energy metabolism and depletion of high energy phosphate compounds; increased superoxide anion levels and a reduced antioxidant capacity; vasoconstriction of arteries; changes in red blood cell (RBC) structure, function and metabolism; alterations in plasma levels of substances involved in coagulation and fibrinolysis and increased endothelin-1 levels and N-terminal-pro brain natriuretic peptide levels. Based on these findings the proposed mechanisms are: endothelial injury followed by thrombosis, increased metabolism leading to energy depletion and ischemia, oxidative stress causing cellular damage, coronary artery spasm leading to myocardial ischemia and diminished ability of RBCs to transfer oxygen resulting in myocardial ischemia. Conclusions There is no evidence for a single mechanism responsible for 5-FU-induced cardiotoxicity, and the underlying mechanisms might be multifactorial. Further research is needed to elucidate the pathogenesis of this side effect.
Collapse
Affiliation(s)
- Anne Polk
- Departments of Cardiology, Herlev Hospital, University of Copenhagen, Herlev Ringvej 75, DK-2730 Herlev, Denmark.
| | | | | | | |
Collapse
|
228
|
Chun JN, Cho M, So I, Jeon JH. The protective effects of Schisandra chinensis fruit extract and its lignans against cardiovascular disease: A review of the molecular mechanisms. Fitoterapia 2014; 97:224-33. [DOI: 10.1016/j.fitote.2014.06.014] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 06/19/2014] [Accepted: 06/20/2014] [Indexed: 02/06/2023]
|
229
|
Herman EH, Hasinoff BB, Steiner R, Lipshultz SE. A review of the preclinical development of dexrazoxane. PROGRESS IN PEDIATRIC CARDIOLOGY 2014. [DOI: 10.1016/j.ppedcard.2014.09.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
230
|
Beneš H, Vuong MK, Boerma M, McElhanon KE, Siegel ER, Singh SP. Protection from oxidative and electrophilic stress in the Gsta4-null mouse heart. Cardiovasc Toxicol 2014; 13:347-56. [PMID: 23690225 DOI: 10.1007/s12012-013-9215-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
4-Hydroxynonenal (4-HNE) mediates many pathological effects of oxidative and electrophilic stress and signals to activate cytoprotective gene expression regulated by NF-E2-related factor 2 (Nrf2). By exhibiting very high levels of 4-HNE-conjugating activity, the murine glutathione transferase alpha 4 (GSTA4-4) helps regulate cellular 4-HNE levels. To examine the role of 4-HNE in vivo, we disrupted the murine Gsta4 gene. Gsta4-null mice exhibited no cardiac phenotype under normal conditions and no difference in cardiac 4-HNE level as compared to wild-type mice. We hypothesized that the Nrf2 pathway might contribute an important compensatory mechanism to remove excess cardiac 4-HNE in Gsta4-null mice. Cardiac nuclear extracts from Gsta4-null mice exhibited significantly higher Nrf2 binding to antioxidant response elements. We also observed responses in critical Nrf2 target gene products: elevated Sod2, Cat, and Akr1b7 mRNA levels and significant increases in both cardiac antioxidant and anti-electrophile enzyme activities. Gsta4-null mice were less sensitive and maintained normal cardiac function following chronic doxorubicin treatment, known to increase cardiac 4-HNE levels. Hence, in the absence of GSTA4-4 to modulate both physiological and pathological 4-HNE levels, the adaptive Nrf2 pathway may be primed to contribute to a preconditioned cardiac phenotype in the Gsta4-null mouse.
Collapse
Affiliation(s)
- Helen Beneš
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | | | | | | | | | | |
Collapse
|
231
|
Nitiss KC, Nitiss JL. Twisting and ironing: doxorubicin cardiotoxicity by mitochondrial DNA damage. Clin Cancer Res 2014; 20:4737-9. [PMID: 24916696 DOI: 10.1158/1078-0432.ccr-14-0821] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Anthracyclines are active clinical agents that have multiple mechanisms of cytotoxicity. Cardiotoxicity by anthracyclines limits the therapeutic potential of these agents, but mechanisms leading to cardiotoxicity remain controversial. Transgenic mice that lack mitochondrial topoisomerase I are hypersensitive to doxorubicin cardiotoxicity, providing support for cardiotoxicity arising from damage of mitochondrial DNA.
Collapse
Affiliation(s)
- Karin C Nitiss
- Biomedical Sciences Department, UIC College of Medicine and Department of Biopharmaceutical Sciences, UIC College of Pharmacy, Rockford, Illinois
| | - John L Nitiss
- Biomedical Sciences Department, UIC College of Medicine and Department of Biopharmaceutical Sciences, UIC College of Pharmacy, Rockford, Illinois.
| |
Collapse
|
232
|
Molecular remodeling of left and right ventricular myocardium in chronic anthracycline cardiotoxicity and post-treatment follow up. PLoS One 2014; 9:e96055. [PMID: 24804796 PMCID: PMC4013127 DOI: 10.1371/journal.pone.0096055] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 04/03/2014] [Indexed: 01/19/2023] Open
Abstract
Chronic anthracycline cardiotoxicity is a serious clinical issue with well characterized functional and histopathological hallmarks. However, molecular determinants of the toxic damage and associated myocardial remodeling remain to be established. Furthermore, details on the different propensity of the left and right ventricle (LV and RV, respectively) to the cardiotoxicity development are unknown. Hence, the aim of the investigation was to study molecular changes associated with remodeling of the LV and RV in chronic anthracycline cardiotoxicity and post-treatment follow up. The cardiotoxicity was induced in rabbits with daunorubicin (3 mg/kg/week for 10 weeks) and animals were sacrificed either at the end of the treatment or after an additional 10 weeks. Daunorubicin induced severe and irreversible cardiotoxicity associated with LV dysfunction and typical morphological alterations, whereas the myocardium of the RV showed only mild changes. Both ventricles also showed different expression of ANP after daunorubicin treatment. Daunorubicin impaired the expression of several sarcomeric proteins in the LV, which was not the case of the RV. In particular, a significant drop was found in titin and thick filament proteins at both mRNA and protein level and this might be connected with persistent LV down-regulation of GATA-4. In addition, the LV was more affected by treatment-induced perturbations in calcium handling proteins. LV cardiomyocytes showed marked up-regulation of desmin after the treatment and vimentin was mainly induced in LV fibroblasts, whereas only weaker changes were observed in the RV. Remodeling of extracellular matrix was almost exclusively found in the LV with particular induction of collagen I and IV. Hence, the present study describes profound molecular remodeling of myocytes, non-myocyte cells and extracellular matrix in response to chronic anthracycline treatment with marked asymmetry between LV and RV.
Collapse
|
233
|
Nrf2 deficiency exaggerates doxorubicin-induced cardiotoxicity and cardiac dysfunction. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014; 2014:748524. [PMID: 24895528 PMCID: PMC4033424 DOI: 10.1155/2014/748524] [Citation(s) in RCA: 129] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 04/10/2014] [Accepted: 04/11/2014] [Indexed: 01/09/2023]
Abstract
The anticancer therapy of doxorubicin (Dox) has been limited by its acute and chronic cardiotoxicity. In addition to a causative role of oxidative stress, autophagy appears to play an important role in the regulation of Dox-induced cardiotoxicity. However, the underlying mechanisms remain unclear. Accordingly, we explored a role of nuclear factor erythroid-2 related factor 2 (Nrf2) in Dox-induced cardiomyopathy with a focus on myocardial oxidative stress and autophagic activity. In wild type (WT) mice, a single intraperitoneal injection of 25 mg/kg Dox rapidly induced cardiomyocyte necrosis and cardiac dysfunction, which were associated with oxidative stress, impaired autophagy, and accumulated polyubiquitinated protein aggregates. However, these Dox-induced adverse effects were exaggerated in Nrf2 knockout (Nrf2(-/-)) mice. In cultured cardiomyocytes, overexpression of Nrf2 increased the steady levels of LC3-II, ameliorated Dox-induced impairment of autophagic flux and accumulation of ubiquitinated protein aggregates, and suppressed Dox-induced cytotoxicity, whereas knockdown of Nrf2 exerted opposite effects. Moreover, the exaggerated adverse effects in Dox-intoxicated Nrf2 depleted cardiomyocytes were dramatically attenuated by forced activation of autophagy via overexpression of autophagy related gene 5 (Atg5). Thus, these results suggest that Nrf2 is likely an endogenous suppressor of Dox-induced cardiotoxicity by controlling both oxidative stress and autophagy in the heart.
Collapse
|
234
|
Maccarinelli F, Gammella E, Asperti M, Regoni M, Biasiotto G, Turco E, Altruda F, Lonardi S, Cornaghi L, Donetti E, Recalcati S, Poli M, Finazzi D, Arosio P, Cairo G. Mice lacking mitochondrial ferritin are more sensitive to doxorubicin-mediated cardiotoxicity. J Mol Med (Berl) 2014; 92:859-69. [PMID: 24728422 PMCID: PMC4118045 DOI: 10.1007/s00109-014-1147-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 02/09/2014] [Accepted: 03/18/2014] [Indexed: 10/26/2022]
Abstract
UNLABELLED Mitochondrial ferritin is a functional ferritin that localizes in the mitochondria. It is expressed in the testis, heart, brain, and cells with active respiratory activity. Its overexpression in cultured cells protected against oxidative damage and reduced cytosolic iron availability. However, no overt phenotype was described in mice with inactivation of the FtMt gene. Here, we used the doxorubicin model of cardiac injury in a novel strain of FtMt-null mice to investigate the antioxidant role of FtMt. These mice did not show any evident phenotype, but after acute treatment to doxorubicin, they showed enhanced mortality and altered heart morphology with fibril disorganization and severe mitochondrial damage. Signs of mitochondrial damage were present also in mock-treated FtMt(-/-) mice. The hearts of saline- and doxorubicin-treated FtMt(-/-) mice had higher thiobarbituric acid reactive substance levels, heme oxygenase 1 expression, and protein oxidation, but did not differ from FtMt(+/+) in the cardiac damage marker B-type natriuretic peptide (BNP), ATP levels, and apoptosis. However, the autophagy marker LC3 was activated. The results show that the absence of FtMt, which is highly expressed in the heart, increases the sensitivity of heart mitochondria to the toxicity of doxorubicin. This study represents the first in vivo evidence of the antioxidant role of FtMt. KEY MESSAGE Mitochondrial ferritin (FtMt) expressed in the heart has a protective antioxidant role. Acute treatment with doxorubicin caused the death of all FtMt(-/-) and only of 60 % FtMt(+/+) mice. The hearts of FtMt(-/-) mice showed fibril disorganization and mitochondrial damage. Markers of oxidative damage and autophagy were increased in FtMt(-/-) hearts. This is the first in vivo evidence of the antioxidant role of FtMt.
Collapse
Affiliation(s)
- Federica Maccarinelli
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123, Brescia, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
235
|
Meyerstein D. Is Measuring OH.Radical Scavenging a Reasonable Measurement of Antioxidant Properties? Isr J Chem 2014. [DOI: 10.1002/ijch.201300123] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
236
|
Gammella E, Maccarinelli F, Buratti P, Recalcati S, Cairo G. The role of iron in anthracycline cardiotoxicity. Front Pharmacol 2014; 5:25. [PMID: 24616701 PMCID: PMC3935484 DOI: 10.3389/fphar.2014.00025] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 02/12/2014] [Indexed: 01/24/2023] Open
Abstract
The clinical use of the antitumor anthracycline Doxorubicin is limited by the risk of severe cardiotoxicity. The mechanisms underlying anthracycline-dependent cardiotoxicity are multiple and remain uncompletely understood, but many observations indicate that interactions with cellular iron metabolism are important. Convincing evidence showing that iron plays a role in Doxorubicin cardiotoxicity is provided by the protecting efficacy of iron chelation in patients and experimental models, and studies showing that iron overload exacerbates the cardiotoxic effects of the drug, but the underlying molecular mechanisms remain to be completely characterized. Since anthracyclines generate reactive oxygen species, increased iron-catalyzed formation of free radicals appears an obvious explanation for the aggravating role of iron in Doxorubicin cardiotoxicity, but antioxidants did not offer protection in clinical settings. Moreover, how the interaction between reactive oxygen species and iron damages heart cells exposed to Doxorubicin is still unclear. This review discusses the pathogenic role of the disruption of iron homeostasis in Doxorubicin-mediated cardiotoxicity in the context of current and future pharmacologic approaches to cardioprotection.
Collapse
Affiliation(s)
- Elena Gammella
- Department of Biomedical Sciences for Health, University of Milano Milano, Italy
| | - Federica Maccarinelli
- Department of Molecular and Translational Medicine, University of Brescia Brescia, Italy
| | - Paolo Buratti
- Department of Biomedical Sciences for Health, University of Milano Milano, Italy
| | - Stefania Recalcati
- Department of Biomedical Sciences for Health, University of Milano Milano, Italy
| | - Gaetano Cairo
- Department of Biomedical Sciences for Health, University of Milano Milano, Italy
| |
Collapse
|
237
|
Potuckova E, Jansova H, Machacek M, Vavrova A, Haskova P, Tichotova L, Richardson V, Kalinowski DS, Richardson DR, Simunek T. Quantitative analysis of the anti-proliferative activity of combinations of selected iron-chelating agents and clinically used anti-neoplastic drugs. PLoS One 2014; 9:e88754. [PMID: 24586383 PMCID: PMC3930662 DOI: 10.1371/journal.pone.0088754] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Accepted: 01/11/2014] [Indexed: 12/11/2022] Open
Abstract
Recent studies have demonstrated that several chelators possess marked potential as potent anti-neoplastic drugs and as agents that can ameliorate some of the adverse effects associated with standard chemotherapy. Anti-cancer treatment employs combinations of several drugs that have different mechanisms of action. However, data regarding the potential interactions between iron chelators and established chemotherapeutics are lacking. Using estrogen receptor-positive MCF-7 breast cancer cells, we explored the combined anti-proliferative potential of four iron chelators, namely: desferrioxamine (DFO), salicylaldehyde isonicotinoyl hydrazone (SIH), (E)-N'-[1-(2-hydroxy-5-nitrophenyl)ethyliden] isonicotinoyl hydrazone (NHAPI), and di-2-pyridylketone 4,4-dimethyl-3-thiosemicarbazone (Dp44mT), plus six selected anti-neoplastic drugs. These six agents are used for breast cancer treatment and include: paclitaxel, 5-fluorouracil, doxorubicin, methotrexate, tamoxifen and 4-hydroperoxycyclophosphamide (an active metabolite of cyclophosphamide). Our quantitative chelator-drug analyses were designed according to the Chou-Talalay method for drug combination assessment. All combinations of these agents yielded concentration-dependent, anti-proliferative effects. The hydrophilic siderophore, DFO, imposed antagonism when used in combination with all six anti-tumor agents and this antagonistic effect increased with increasing dose. Conversely, synergistic interactions were observed with combinations of the lipophilic chelators, NHAPI or Dp44mT, with doxorubicin and also the combinations of SIH, NHAPI or Dp44mT with tamoxifen. The combination of Dp44mT with anti-neoplastic agents was further enhanced following formation of its redox-active iron and especially copper complexes. The most potent combinations of Dp44mT and NHAPI with tamoxifen were confirmed as synergistic using another estrogen receptor-expressing breast cancer cell line, T47D, but not estrogen receptor-negative MDA-MB-231 cells. Furthermore, the synergy of NHAPI and tamoxifen was confirmed using MCF-7 cells by electrical impedance data, a mitochondrial inner membrane potential assay and cell cycle analyses. This is the first systematic investigation to quantitatively assess interactions between Fe chelators and standard chemotherapies using breast cancer cells. These studies are vital for their future clinical development.
Collapse
Affiliation(s)
- Eliska Potuckova
- Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové, Charles University in Prague, Prague, Czech Republic
| | - Hana Jansova
- Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové, Charles University in Prague, Prague, Czech Republic
| | - Miloslav Machacek
- Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové, Charles University in Prague, Prague, Czech Republic
| | - Anna Vavrova
- Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové, Charles University in Prague, Prague, Czech Republic
| | - Pavlina Haskova
- Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové, Charles University in Prague, Prague, Czech Republic
| | - Lucie Tichotova
- Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové, Charles University in Prague, Prague, Czech Republic
| | - Vera Richardson
- Molecular Pharmacology and Pathology Program, Bosch Institute and Department of Pathology, University of Sydney, Sydney, Australia
| | - Danuta S. Kalinowski
- Molecular Pharmacology and Pathology Program, Bosch Institute and Department of Pathology, University of Sydney, Sydney, Australia
| | - Des R. Richardson
- Molecular Pharmacology and Pathology Program, Bosch Institute and Department of Pathology, University of Sydney, Sydney, Australia
- * E-mail: (TS); (DRR)
| | - Tomas Simunek
- Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové, Charles University in Prague, Prague, Czech Republic
- * E-mail: (TS); (DRR)
| |
Collapse
|
238
|
Lagoa R, Gañán C, López-Sánchez C, García-Martínez V, Gutierrez-Merino C. The decrease of NAD(P)H:quinone oxidoreductase 1 activity and increase of ROS production by NADPH oxidases are early biomarkers in doxorubicin cardiotoxicity. Biomarkers 2014; 19:142-53. [PMID: 24506563 DOI: 10.3109/1354750x.2014.885084] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
CONTEXT Doxorubicin cardiotoxicity displays a complex and multifactorial progression. OBJECTIVE Identify early biochemical mechanisms leading to a sustained imbalance of cellular bioenergetics. METHODS Measurements of the temporal evolution of selected biochemical markers after treatment of rats with doxorubicin (20 mg/kg body weight). RESULTS Doxorubicin treatment increased lipid oxidation, catalase activity and production of H₂O₂ by Nox-NADPH oxidases, and down-regulated NAD(P)H quinone oxidoreductase-1 prior eliciting changes in reduced glutathione, protein carbonyls and protein nitrotyrosines. Alterations of mitochondrial and myofibrillar bioenergetics biomarkers were detected only after this oxidative imbalance was established. CONCLUSIONS NAD(P)H quinone oxidoreductase-1 activity and increase of hydrogen peroxide production by NADPH oxidases are early biomarkers in doxorubicin cardiotoxicity.
Collapse
Affiliation(s)
- Ricardo Lagoa
- ESTG-Polytechnic Institute of Leiria , Leiria , Portugal
| | | | | | | | | |
Collapse
|
239
|
Tranoy-Opalinski I, Legigan T, Barat R, Clarhaut J, Thomas M, Renoux B, Papot S. β-Glucuronidase-responsive prodrugs for selective cancer chemotherapy: an update. Eur J Med Chem 2014; 74:302-13. [PMID: 24480360 DOI: 10.1016/j.ejmech.2013.12.045] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 12/22/2013] [Accepted: 12/23/2013] [Indexed: 02/07/2023]
Abstract
The design of novel antitumor agents allowing the destruction of malignant cells while sparing healthy tissues is one of the major challenges in medicinal chemistry. In this context, the use of non-toxic prodrugs programmed to be selectively activated by beta-glucuronidase present at high concentration in the microenvironment of most solid tumors has attracted considerable attention. This review summarizes the major progresses that have been realized in this field over the past ten years. This includes the new prodrugs that have been designed to target a wide variety of anticancer drugs, the prodrugs employed in the course of a combined therapy, the dendritic glucuronide prodrugs and the concept of β-glucuronidase-responsive albumin binding prodrugs.
Collapse
Affiliation(s)
- Isabelle Tranoy-Opalinski
- Université de Poitiers, UMR-CNRS 7285, Institut de Chimie des Milieux et des Matériaux de Poitiers (IC2MP), Groupe "Systèmes Moléculaires Programmés", 4 rue Michel Brunet, 86022 Poitiers, France
| | - Thibaut Legigan
- Université de Poitiers, UMR-CNRS 7285, Institut de Chimie des Milieux et des Matériaux de Poitiers (IC2MP), Groupe "Systèmes Moléculaires Programmés", 4 rue Michel Brunet, 86022 Poitiers, France
| | - Romain Barat
- Université de Poitiers, UMR-CNRS 7285, Institut de Chimie des Milieux et des Matériaux de Poitiers (IC2MP), Groupe "Systèmes Moléculaires Programmés", 4 rue Michel Brunet, 86022 Poitiers, France
| | - Jonathan Clarhaut
- Université de Poitiers, UMR-CNRS 7285, Institut de Chimie des Milieux et des Matériaux de Poitiers (IC2MP), Groupe "Systèmes Moléculaires Programmés", 4 rue Michel Brunet, 86022 Poitiers, France; INSERM CIC 0802, CHU de Poitiers, 2 rue de la Milétrie, 86021 Poitiers, France
| | - Mikaël Thomas
- Université de Poitiers, UMR-CNRS 7285, Institut de Chimie des Milieux et des Matériaux de Poitiers (IC2MP), Groupe "Systèmes Moléculaires Programmés", 4 rue Michel Brunet, 86022 Poitiers, France
| | - Brigitte Renoux
- Université de Poitiers, UMR-CNRS 7285, Institut de Chimie des Milieux et des Matériaux de Poitiers (IC2MP), Groupe "Systèmes Moléculaires Programmés", 4 rue Michel Brunet, 86022 Poitiers, France
| | - Sébastien Papot
- Université de Poitiers, UMR-CNRS 7285, Institut de Chimie des Milieux et des Matériaux de Poitiers (IC2MP), Groupe "Systèmes Moléculaires Programmés", 4 rue Michel Brunet, 86022 Poitiers, France.
| |
Collapse
|
240
|
|
241
|
Corradi F, Paolini L, De Caterina R. Ranolazine in the prevention of anthracycline cardiotoxicity. Pharmacol Res 2014; 79:88-102. [DOI: 10.1016/j.phrs.2013.11.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2013] [Revised: 11/06/2013] [Accepted: 11/06/2013] [Indexed: 12/19/2022]
|
242
|
Gao S, Li H, Cai Y, Ye JT, Liu ZP, Lu J, Huang XY, Feng XJ, Gao H, Chen SR, Li M, Liu PQ. Mitochondrial binding of α-enolase stabilizes mitochondrial membrane: Its role in doxorubicin-induced cardiomyocyte apoptosis. Arch Biochem Biophys 2014; 542:46-55. [DOI: 10.1016/j.abb.2013.12.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2013] [Revised: 12/05/2013] [Accepted: 12/08/2013] [Indexed: 12/20/2022]
|
243
|
Vavrova A, Jansova H, Mackova E, Machacek M, Haskova P, Tichotova L, Sterba M, Simunek T. Catalytic inhibitors of topoisomerase II differently modulate the toxicity of anthracyclines in cardiac and cancer cells. PLoS One 2013; 8:e76676. [PMID: 24116135 PMCID: PMC3792022 DOI: 10.1371/journal.pone.0076676] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 08/25/2013] [Indexed: 01/09/2023] Open
Abstract
Anthracyclines (such as doxorubicin or daunorubicin) are among the most effective anticancer drugs, but their usefulness is hampered by the risk of irreversible cardiotoxicity. Dexrazoxane (ICRF-187) is the only clinically approved cardioprotective agent against anthracycline cardiotoxicity. Its activity has traditionally been attributed to the iron-chelating effects of its metabolite with subsequent protection from oxidative stress. However, dexrazoxane is also a catalytic inhibitor of topoisomerase II (TOP2). Therefore, we examined whether dexrazoxane and two other TOP2 catalytic inhibitors, namely sobuzoxane (MST-16) and merbarone, protect cardiomyocytes from anthracycline toxicity and assessed their effects on anthracycline antineoplastic efficacy. Dexrazoxane and two other TOP2 inhibitors protected isolated neonatal rat cardiomyocytes against toxicity induced by both doxorubicin and daunorubicin. However, none of the TOP2 inhibitors significantly protected cardiomyocytes in a model of hydrogen peroxide-induced oxidative injury. In contrast, the catalytic inhibitors did not compromise the antiproliferative effects of the anthracyclines in the HL-60 leukemic cell line; instead, synergistic interactions were mostly observed. Additionally, anthracycline-induced caspase activation was differentially modulated by the TOP2 inhibitors in cardiac and cancer cells. Whereas dexrazoxane was upon hydrolysis able to significantly chelate intracellular labile iron ions, no such effect was noted for either sobuzoxane or merbarone. In conclusion, our data indicate that dexrazoxane may protect cardiomyocytes via its catalytic TOP2 inhibitory activity rather than iron-chelation activity. The differential expression and/or regulation of TOP2 isoforms in cardiac and cancer cells by catalytic inhibitors may be responsible for the selective modulation of anthracycline action observed.
Collapse
Affiliation(s)
- Anna Vavrova
- Department of Biochemical Sciences, Charles University in Prague, Faculty of Pharmacy in Hradec Kralove, Hradec Kralove, Czech Republic
| | - Hana Jansova
- Department of Biochemical Sciences, Charles University in Prague, Faculty of Pharmacy in Hradec Kralove, Hradec Kralove, Czech Republic
| | - Eliska Mackova
- Department of Biochemical Sciences, Charles University in Prague, Faculty of Pharmacy in Hradec Kralove, Hradec Kralove, Czech Republic
| | - Miloslav Machacek
- Department of Biochemical Sciences, Charles University in Prague, Faculty of Pharmacy in Hradec Kralove, Hradec Kralove, Czech Republic
| | - Pavlina Haskova
- Department of Biochemical Sciences, Charles University in Prague, Faculty of Pharmacy in Hradec Kralove, Hradec Kralove, Czech Republic
| | - Lucie Tichotova
- Department of Biochemical Sciences, Charles University in Prague, Faculty of Pharmacy in Hradec Kralove, Hradec Kralove, Czech Republic
| | - Martin Sterba
- Department of Pharmacology, Charles University in Prague, Faculty of Medicine in Hradec Kralove, Hradec Kralove, Czech Republic
| | - Tomas Simunek
- Department of Biochemical Sciences, Charles University in Prague, Faculty of Pharmacy in Hradec Kralove, Hradec Kralove, Czech Republic
| |
Collapse
|
244
|
Abstract
Modern cancer therapies are highly effective in the treatment of various malignancies, but their use is limited by the potential for cardiotoxicity. The most frequent and typical clinical manifestation of cardiotoxicity is left ventricular dysfunction, induced not only by cytotoxic conventional cancer therapy like anthracyclines, but also by new antitumor targeted therapy such as trastuzumab. The current standard for monitoring cardiac function, based on periodic assessment of left ventricular ejection fraction detects cardiotoxicity only when a functional impairment has already occurred, precluding any chance of preventing its development. A novel approach, based on the use of cardiac biomarkers has emerged in the last decade, resulting in a cost-effective diagnostic tool for early, real-time identification, assessment and monitoring of cardiotoxicity. In particular, prophylactic treatment with enalapril in patients with an early increase in troponin after chemotherapy has been shown to be very effective in preventing left ventricular dysfunction and associated cardiac events. In patients developing cancer treatment induced-cardiomyopathy, complete left ventricular ejection fraction recovery and a reduction of cardiac events may be achieved only when left ventricular dysfunction is detected early after the end of cancer treatment and treatment with angiotensin-converting enzyme inhibitors, possibly in combination with beta-blockers, is promptly initiated.
Collapse
|
245
|
Jirkovský E, Lenčová-Popelová O, Hroch M, Adamcová M, Mazurová Y, Vávrová J, Mičuda S, Šimůnek T, Geršl V, Štěrba M. Early and delayed cardioprotective intervention with dexrazoxane each show different potential for prevention of chronic anthracycline cardiotoxicity in rabbits. Toxicology 2013; 311:191-204. [PMID: 23831762 DOI: 10.1016/j.tox.2013.06.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Revised: 06/25/2013] [Accepted: 06/26/2013] [Indexed: 10/26/2022]
Abstract
Despite incomplete understanding to its mechanism of action, dexrazoxane (DEX) is still the only clearly effective cardioprotectant against chronic anthracycline (ANT) cardiotoxicity. However, its clinical use is currently restricted to patients exceeding significant ANT cumulative dose (300mg/m(2)), although each ANT cycle may induce certain potentially irreversible myocardial damage. Therefore, the aim of this study was to compare early and delayed DEX intervention against chronic ANT cardiotoxicity and study the molecular events involved. The cardiotoxicity was induced in rabbits with daunorubicin (DAU; 3mg/kg/week for 10 weeks); DEX (60mg/kg) was administered either before the 1st or 7th DAU dose (i.e. after ≈300mg/m(2) cumulative dose). While both DEX administration schedules prevented DAU-induced premature deaths and severe congestive heart failure, only the early intervention completely prevented the left ventricular dysfunction, myocardial morphological changes and mitochondrial damage. Further molecular analyses did not support the assumption that DEX cardioprotection is based and directly proportional to protection from DAU-induced oxidative damage and/or deletions in mtDNA. Nevertheless, DAU induced significant up-regulation of heme oxygenase 1 pathway while heme synthesis was inversely regulated and both changes were schedule-of-administration preventable by DEX. Early and delayed DEX interventions also differed in ability to prevent DAU-induced down-regulation of expression of mitochondrial proteins encoded by both nuclear and mitochondrial genome. Hence, the present functional, morphological as well as the molecular data highlights the enormous cardioprotective effects of DEX and provides novel insights into the molecular events involved. Furthermore, the data suggests that currently recommended delayed intervention may not be able to take advantage of the full cardioprotective potential of the drug.
Collapse
Affiliation(s)
- Eduard Jirkovský
- Department of Pharmacology, Faculty of Medicine in Hradec Králové, Charles University in Prague, Šimkova 870, Hradec Králové 500 38, Czech Republic
| | | | | | | | | | | | | | | | | | | |
Collapse
|
246
|
Novel aspects of ROS signalling in heart failure. Basic Res Cardiol 2013; 108:359. [PMID: 23740217 DOI: 10.1007/s00395-013-0359-8] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 05/03/2013] [Accepted: 05/05/2013] [Indexed: 12/20/2022]
Abstract
Heart failure and many of the conditions that predispose to heart failure are associated with oxidative stress. This is considered to be important in the pathophysiology of the condition but clinical trials of antioxidant approaches to prevent cardiovascular morbidity and mortality have been unsuccessful. Part of the reason for this may be the failure to appreciate the complexity of the effects of reactive oxygen species. At one extreme, excessive oxidative stress damages membranes, proteins and DNA but lower levels of reactive oxygen species may exert much more subtle and specific regulatory effects (termed redox signalling), even on physiological signalling pathways. In this article, we review our current understanding of the roles of such redox signalling pathways in the pathophysiology of heart failure, including effects on cardiomyocyte hypertrophy signalling, excitation-contraction coupling, arrhythmia, cell viability and energetics. Reactive oxygen species generated by NADPH oxidase proteins appear to be especially important in redox signalling. The delineation of specific redox-sensitive pathways and mechanisms that contribute to different components of the failing heart phenotype may facilitate the development of newer targeted therapies as opposed to the failed general antioxidant approaches of the past.
Collapse
|
247
|
An T, Zhang Y, Huang Y, Zhang R, Yin S, Guo X, Wang Y, Zou C, Wei B, Lv R, Zhou Q, Zhang J. Neuregulin-1 protects against doxorubicin-induced apoptosis in cardiomyocytes through an Akt-dependent pathway. Physiol Res 2013; 62:379-85. [PMID: 23590603 DOI: 10.33549/physiolres.932516] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
In previous studies, it has been shown that recombinant human neuregulin-1(rhNRG-1) is capable of improving the survival rate in animal models of doxorubicin (DOX)-induced cardiomyopathy; however, the underlying mechanism of this phenomenon remains unknown. In this study, the role of rhNRG-1 in attenuating doxorubicin-induce apoptosis is confirmed. Neonatal rat ventricular myocytes (NRVMs) were subjected to various treatments, in order to both induce apoptosis and determine the effects of rhNRG-1 on the process. Activation of apoptosis was determined by observing increases in the protein levels of classic apoptosis markers (including cleaved caspase-3, cytochrome c, Bcl-2, BAX and terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling (TUNEL) staining). The activation of Akt was detected by means of western blot analysis. The study results showed that doxorubicin increased the number of TUNEL positive cells, as well as the protein levels of cleaved caspase-3 and cytochrome c, and reduced the ratio of Bcl-2/Bax. However, all of these effects were markedly antagonized by pretreament with rhNRG-1. It was then further demonstrated that the effects of rhNRG-1 could be blocked by the phosphoinositole-3-kinase inhibitor LY294002, indicating the involvement of the Akt process in mediating the process. RhNRG-1 is a potent inhibitor of doxorubicin-induced apoptosis, which acts through the PI3K-Akt pathway. RhNRG-1 is a novel therapeutic drug which may be effective in preventing further damage from occurring in DOX-induced damaged myocardium.
Collapse
Affiliation(s)
- T An
- Heart Failure Center, Cardiovascular Institute and Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
248
|
Kovarikova P, Pasakova-Vrbatova I, Vavrova A, Stariat J, Klimes J, Simunek T. Development of LC–MS/MS method for the simultaneous analysis of the cardioprotective drug dexrazoxane and its metabolite ADR-925 in isolated cardiomyocytes and cell culture medium. J Pharm Biomed Anal 2013; 76:243-51. [DOI: 10.1016/j.jpba.2012.12.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Revised: 12/08/2012] [Accepted: 12/10/2012] [Indexed: 11/28/2022]
|
249
|
Abstract
Mitochondria are primarily responsible for providing the contracting cardiac myocyte with a continuous supply of ATP. However, mitochondria can rapidly change into death-promoting organelles. In response to changes in the intracellular environment, mitochondria become producers of excessive reactive oxygen species and release prodeath proteins, resulting in disrupted ATP synthesis and activation of cell death pathways. Interestingly, cells have developed a defense mechanism against aberrant mitochondria that can cause harm to the cell. This mechanism involves selective sequestration and subsequent degradation of the dysfunctional mitochondrion before it causes activation of cell death. Induction of mitochondrial autophagy, or mitophagy, results in selective clearance of damaged mitochondria in cells. In response to stress such as ischemia/reperfusion, prosurvival and prodeath pathways are concomitantly activated in cardiac myocytes. Thus, there is a delicate balance between life and death in the myocytes during stress, and the final outcome depends on the complex cross-talk between these pathways. Mitophagy functions as an early cardioprotective response, favoring adaptation to stress by removing damaged mitochondria. In contrast, increased oxidative stress and apoptotic proteases can inactivate mitophagy, allowing for the execution of cell death. Herein, we discuss the importance of mitochondria and mitophagy in cardiovascular health and disease and provide a review of our current understanding of how these processes are regulated.
Collapse
Affiliation(s)
- Dieter A Kubli
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | | |
Collapse
|
250
|
Epirubicin loaded with propylene glycol liposomes significantly overcomes multidrug resistance in breast cancer. Cancer Lett 2012. [PMID: 23186833 DOI: 10.1016/j.canlet.2012.11.031] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Multidrug resistance (MDR) is one of the major reasons for the failure of cancer chemotherapy. A newly reported liposome carrier, propylene glycol liposomes (EPI-PG-liposomes) were made to load epirubicin (EPI) which enhanced EPI absorption in MDR tumor cells to overcome the drug resistance. MDA-MB 435 and their mutant resistant (MDA-MB 435/ADR) cells were used to examine the cellular uptake and P-gp function in vitro for EPI-PG-liposomes by fluorescence microscopy and FCM, respectively. Mammary tumor model was also established to investigate the tumor growth inhibition and pharmacodynamics of EPI-PG-liposomes in vivo. Morphology evaluation showed that EPI-PG-liposomes had a homogeneous spherical shape with an average diameter of 182 nm. Based on cell viability assay, fluorescent microscopy examination, and EPI uptake assay, EPI-PG-liposomes exhibited an effective growth inhibition not only in MDA-MB-435 cells, but also in MDA-MB 435/ADR cells. EPI-PG-liposomes have high permeability not only on tumor cell membrane, but also on cell nucleus membrane. P-gp function assay showed that the anticancer action of EPI-PG-liposomes was not related to P-gp efflux pump, suggesting that PG-liposomes would not affect the normal physiological functions of membrane proteins. EPI-PG-liposomes also showed a better antitumor efficacy compared to EPI solution alone. With high entrapment efficiency, spherical morphology and effective inhibition on MDR cancer cells, EPI-PG-liposomes may represent a better chemotherapeutic vectors for cancer targeted therapy.
Collapse
|