301
|
Ramipril restores PPARβ/δ and PPARγ expressions and reduces cardiac NADPH oxidase but fails to restore cardiac function and accompanied myosin heavy chain ratio shift in severe anthracycline-induced cardiomyopathy in rat. Eur J Pharmacol 2016; 791:244-253. [DOI: 10.1016/j.ejphar.2016.08.040] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 08/31/2016] [Accepted: 08/31/2016] [Indexed: 01/25/2023]
|
302
|
Shahidi F, de Camargo AC. Tocopherols and Tocotrienols in Common and Emerging Dietary Sources: Occurrence, Applications, and Health Benefits. Int J Mol Sci 2016; 17:E1745. [PMID: 27775605 PMCID: PMC5085773 DOI: 10.3390/ijms17101745] [Citation(s) in RCA: 179] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 10/05/2016] [Accepted: 10/13/2016] [Indexed: 12/11/2022] Open
Abstract
Edible oils are the major natural dietary sources of tocopherols and tocotrienols, collectively known as tocols. Plant foods with low lipid content usually have negligible quantities of tocols. However, seeds and other plant food processing by-products may serve as alternative sources of edible oils with considerable contents of tocopherols and tocotrienols. Tocopherols are among the most important lipid-soluble antioxidants in food as well as in human and animal tissues. Tocopherols are found in lipid-rich regions of cells (e.g., mitochondrial membranes), fat depots, and lipoproteins such as low-density lipoprotein cholesterol. Their health benefits may also be explained by regulation of gene expression, signal transduction, and modulation of cell functions. Potential health benefits of tocols include prevention of certain types of cancer, heart disease, and other chronic ailments. Although deficiencies of tocopherol are uncommon, a continuous intake from common and novel dietary sources of tocopherols and tocotrienols is advantageous. Thus, this contribution will focus on the relevant literature on common and emerging edible oils as a source of tocols. Potential application and health effects as well as the impact of new cultivars as sources of edible oils and their processing discards are presented. Future trends and drawbacks are also briefly covered.
Collapse
Affiliation(s)
- Fereidoon Shahidi
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, NL A1B 3X9, Canada.
| | - Adriano Costa de Camargo
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, NL A1B 3X9, Canada.
- Department of Agri-Food Industry, Food & Nutrition, "Luiz de Queiroz" College of Agriculture, University of São Paulo, Piracicaba 13418-900, Brazil.
| |
Collapse
|
303
|
Vuda M, Kamath A. Drug induced mitochondrial dysfunction: Mechanisms and adverse clinical consequences. Mitochondrion 2016; 31:63-74. [PMID: 27771494 DOI: 10.1016/j.mito.2016.10.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 09/24/2016] [Accepted: 10/17/2016] [Indexed: 01/12/2023]
Abstract
Several commonly used medications impair mitochondrial function resulting in adverse effects or toxicities. Drug induced mitochondrial dysfunction may be a consequence of increased production of reactive oxygen species, altered mitochondrial permeability transition, impaired mitochondrial respiration, mitochondrial DNA damage or inhibition of beta-oxidation of fatty acids. The clinical manifestation depends on the specific drug and its effect on mitochondria. Given the ubiquitous presence of mitochondria and its central role in cellular metabolism, drug-mitochondrial interactions may manifest clinically as hepatotoxicity, enteropathy, myelosuppression, lipodystrophy syndrome or neuropsychiatric adverse effects, to name a few. The current review focuses on specific drug groups which adversely affect mitochondria, the mechanisms involved and the clinical consequences based on the data available from experimental and clinical studies. Knowledge of these adverse drug-mitochondrial interactions may help the clinicians foresee potential issues in individual patients, prevent adverse drug reactions or alter drug regimens to enhance patient safety.
Collapse
Affiliation(s)
| | - Ashwin Kamath
- Department of Pharmacology, Kasturba Medical College, Manipal University, Mangalore, India.
| |
Collapse
|
304
|
Mitoxantrone-Surfactant Interactions: A Physicochemical Overview. Molecules 2016; 21:molecules21101356. [PMID: 27754390 PMCID: PMC6273455 DOI: 10.3390/molecules21101356] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 09/29/2016] [Accepted: 10/01/2016] [Indexed: 11/17/2022] Open
Abstract
Mitoxantrone is a synthetic anticancer drug used clinically in the treatment of different types of cancer. It was developed as a doxorubicin analogue in a program to find drugs with improved antitumor activity and decreased cardiotoxicity compared with the anthracyclines. As the cell membrane is the first barrier encountered by anticancer drugs before reaching the DNA sites inside the cells and as surfactant micelles are known as simple model systems for biological membranes, the drugs-surfactant interaction has been the subject of great research interest. Further, quantitative understanding of the interactions of drugs with biomimicking structures like surfactant micelles may provide helpful information for the control of physicochemical properties and bioactivities of encapsulated drugs in order to design better delivery systems with possible biomedical applications. The present review describes the physicochemical aspects of the interactions between the anticancer drug mitoxantrone and different surfactants. Mitoxantrone-micelle binding constants, partitions coefficient of the drug between aqueous and micellar phases and the corresponding Gibbs free energy for the above processes, and the probable location of drug molecules in the micelles are discussed.
Collapse
|
305
|
Swiech O, Majdecki M, Debinski A, Krzak A, Stępkowski TM, Wójciuk G, Kruszewski M, Bilewicz R. Competition between self-inclusion and drug binding explains the pH dependence of the cyclodextrin drug carrier - molecular modelling and electrochemistry studies. NANOSCALE 2016; 8:16733-16742. [PMID: 27714103 DOI: 10.1039/c6nr05833g] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
A non-toxic lipoic acid derivative of β cyclodextrin (βCDLip) with an electron-rich aromatic linker was studied as a carrier for the drug doxorubicin with the aim of decreasing the toxic side effects of this drug. The modified cyclodextrin strengthened the drug binding and differentiated the complex-forming ability with dependence on pH. The stability constants of the complexes were evaluated by voltammetry and spectrofluorometry. Molecular modelling provided deeper insight into the nature of the ligand structure itself and the drug-ligand interactions, showing the different contributions of the self-inclusion of the ligand substituent at different pH values. As a result, the modes of interaction of βCDLip with the drug and factors affecting the stabilities of the complex under the pH conditions of healthy and tumour cells could be discovered and explained.
Collapse
Affiliation(s)
- Olga Swiech
- University of Warsaw, Faculty of Chemistry, Pasteura 1 02-093 Warsaw, Poland.
| | - Maciej Majdecki
- University of Warsaw, Faculty of Chemistry, Pasteura 1 02-093 Warsaw, Poland.
| | - Aleksander Debinski
- University of Warsaw, Faculty of Chemistry, Pasteura 1 02-093 Warsaw, Poland.
| | - Agata Krzak
- University of Warsaw, Faculty of Chemistry, Pasteura 1 02-093 Warsaw, Poland.
| | - Tomasz M Stępkowski
- Department of Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Dorodna 16, Warsaw, 03-195, Poland.
| | - Grzegorz Wójciuk
- Department of Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Dorodna 16, Warsaw, 03-195, Poland.
| | - Marcin Kruszewski
- Faculty of Medicine, University of Information Technology and Management in Rzeszów, ul. Sucharskiego 2, Rzeszów, 35-225, Poland. and Department of Molecular Biology and Translational Research, Institute of Rural Health, Jaczewskiego 2, Lublin, 20-090, Poland.
| | - Renata Bilewicz
- University of Warsaw, Faculty of Chemistry, Pasteura 1 02-093 Warsaw, Poland.
| |
Collapse
|
306
|
Huang PC, Kuo WW, Shen CY, Chen YF, Lin YM, Ho TJ, Padma VV, Lo JF, Huang CY, Huang CY. Anthocyanin Attenuates Doxorubicin-Induced Cardiomyotoxicity via Estrogen Receptor-α/β and Stabilizes HSF1 to Inhibit the IGF-IIR Apoptotic Pathway. Int J Mol Sci 2016; 17:E1588. [PMID: 27657062 PMCID: PMC5037853 DOI: 10.3390/ijms17091588] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 08/23/2016] [Accepted: 09/13/2016] [Indexed: 11/16/2022] Open
Abstract
Doxorubicin (Dox) is extensively used for chemotherapy in different types of cancer, but its use is limited to because of its cardiotoxicity. Our previous studies found that doxorubicin-induced insulin-like growth factor II receptor (IGF-IIR) accumulation causes cardiomyocytes apoptosis via down-regulation of HSF1 pathway. In these studies, we demonstrated a new mechanism through which anthocyanin protects cardiomyoblast cells against doxorubicin-induced injury. We found that anthocyanin decreased IGF-IIR expression via estrogen receptors and stabilized heat shock factor 1 (HSF1) to inhibit caspase 3 activation and apoptosis of cardiomyocytes. Therefore, the phytoestrogen from plants has been considered as another potential treatment for heart failure. It has been reported that the natural compound anthocyanin (ACN) has the ability to reduce the risk of cardiovascular disease (CVD). Here, we demonstrated that anthocyanin acts as a cardioprotective drug against doxorubicin-induced heart failure by attenuating cardiac apoptosis via estrogen receptors to stabilize HSF1 expression and down-regulated IGF-IIR-induced cardiomyocyte apoptosis.
Collapse
Affiliation(s)
- Pei-Chen Huang
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung 40402, Taiwan.
- Department of Obstetrics and Gynecology, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan.
| | - Wei-Wen Kuo
- Department of Biological Science and Technology, China Medical University, Taichung 40402, Taiwan.
| | - Chia-Yao Shen
- Department of Nursing, Mei Ho University, Pingguang Road, Pingtung 91202, Taiwan.
| | - Yu-Feng Chen
- Section of Cardiology, Yuan Rung Hospital, Yuanlin 51045, Taiwan.
| | - Yueh-Min Lin
- Department of Pathology, Changhua Christian Hospital, Changhua 500, Taiwan.
- Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli 35664, Taiwan.
| | - Tsung-Jung Ho
- Chinese Medicine Department, China Medical University Beigang Hospital, Taichung 40402, Taiwan.
| | - V Vijaya Padma
- Department of Biotechnology, Bharathiar University, Coimbatore 641046, India.
| | - Jeng-Fan Lo
- Institute of Oral Biology, National Yang-Ming University, Taipei 11221, Taiwan.
| | - Chih-Yang Huang
- Translation Research Core, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan.
| | - Chih-Yang Huang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung 40402, Taiwan.
- Graduate Institute of Chinese Medical Science, China Medical University, Hsueh-Shih Road, Taichung 40402, Taiwan.
- Department of Health and Nutrition Biotechnology, Asia University, Taichung 40402, Taiwan.
| |
Collapse
|
307
|
Li J, Falcone ER, Holstein SA, Anderson AC, Wright DL, Wiemer AJ. Novel α-substituted tropolones promote potent and selective caspase-dependent leukemia cell apoptosis. Pharmacol Res 2016; 113:438-448. [PMID: 27663262 DOI: 10.1016/j.phrs.2016.09.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 09/07/2016] [Accepted: 09/19/2016] [Indexed: 10/21/2022]
Abstract
Tropolones, such as β-thujaplicin, are small lead-like natural products that possess a variety of biological activities. While the β-substituted natural products and their synthetic analogs are potent inhibitors of human cancer cell growth, less is known about their α-substituted counterparts. Recently, we synthesized a series of α-substituted tropolones including 2-hydroxy-7-(naphthalen-2-yl)cyclohepta-2,4,6-trien-1-one (α-naphthyl tropolone). Here, we evaluate the antiproliferative mechanisms of α-naphthyl tropolone and the related α-benzodioxinyl analog. The α-substituted tropolones inhibit growth of lymphocytic leukemia cells, but not healthy blood cells, with nanomolar potency. Treatment of leukemia cell lines with the tropolone dose-dependently induces apoptosis as judged by staining with annexin V and propidium iodide and Western blot analysis of cleaved caspase 3 and 7. Moreover, pre-treatment of cells with the caspase inhibitor Z-VAD-FMK inhibited the apoptotic effects of the tropolone in two lymphocytic lines. Caspase inhibition also blocked elevated histone acetylation caused by the tropolone, indicating that its effects on histone acetylation are potentiated by caspases. In contrast, α-naphthyl tropolone upregulated p53 expression and phosphorylation of Akt and mTOR in a manner that was not rescued by caspase inhibition. The effects of tropolone were blocked by co-incubation with high levels of free extracellular iron but not by pre-loading with iron. Additionally, dose and time dependent reduction in ex vivo viability of cells from leukemia patients was observed. Taken together, we demonstrate that α-substituted tropolones upregulate DNA damage repair pathways leading to caspase-dependent apoptosis in malignant lymphocytes.
Collapse
Affiliation(s)
- Jin Li
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, United States
| | - Eric R Falcone
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, United States
| | - Sarah A Holstein
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY, United States
| | - Amy C Anderson
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, United States
| | - Dennis L Wright
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, United States
| | - Andrew J Wiemer
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, United States; Institute for Systems Genomics, University of Connecticut, Storrs, CT, United States.
| |
Collapse
|
308
|
Liu ST, Huang SM, Ho CL, Yen LC, Huang CJ, Lin WS, Chan JYH. The regulatory mechanisms of myogenin expression in doxorubicin-treated rat cardiomyocytes. Oncotarget 2016; 6:37443-57. [PMID: 26452256 PMCID: PMC4741940 DOI: 10.18632/oncotarget.5338] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 09/25/2015] [Indexed: 01/03/2023] Open
Abstract
Doxorubicin, an anthracycline antibiotic, has been used as an anti-neoplastic drug for almost 60 years. However, the mechanism(s) by which anthracyclines cause irreversible myocardial injury remains unclear. In order to delineate possible molecular signals involved in the myocardial toxicity, we assessed candidate genes using mRNA expression profiling in the doxorubicin-treated rat cardiomyocyte H9c2 cell line. In the study, it was confirmed that myogenin, an important transcriptional factor for muscle terminal differentiation, was significantly reduced by doxorubicin in a dose-dependent manner using both RT-PCR and western blot analyses. Also, it was identified that the doxorubicin-reduced myogenin gene level could not be rescued by most cardio-protectants. Furthermore, it was demonstrated how the signaling of the decreased myogenin expression by doxorubicin was altered at the transcriptional, post-transcriptional and translational levels. Based on these findings, a working model was proposed for relieving doxorubicin-associated myocardial toxicity by down-regulating miR-328 expression and increasing voltage-gated calcium channel β1 expression, which is a repressor of myogenin gene regulation. In summary, this study provides several lines of evidence indicating that myogenin is the target for doxorubicin-induced cardio-toxicity and a novel therapeutic strategy for doxorubicin clinical applications based on the regulatory mechanisms of myogenin expression.
Collapse
Affiliation(s)
- Shu-Ting Liu
- Department of Biochemistry, National Defense Medical Center, Taipei 114, Taiwan, Republic of China.,Department of Medicine, Division of Hematology/Oncology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan, Republic of China
| | - Shih-Ming Huang
- Department of Biochemistry, National Defense Medical Center, Taipei 114, Taiwan, Republic of China
| | - Ching-Liang Ho
- Department of Medicine, Division of Hematology/Oncology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan, Republic of China
| | - Li-Chen Yen
- Department of Biochemistry, National Defense Medical Center, Taipei 114, Taiwan, Republic of China
| | - Chi-Jung Huang
- Department of Biochemistry, National Defense Medical Center, Taipei 114, Taiwan, Republic of China.,Department of Medical Research, Cathay General Hospital, New Taipei City 221, Taiwan, Republic of China
| | - Wei-Shiang Lin
- Division of Cardiology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei City 114, Taiwan, Republic of China
| | - James Yi-Hsin Chan
- Department of Microbiology and Immunology, National Defense Medical Center, Taipei 114, Taiwan, Republic of China.,Department of Family and Community Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan, Republic of China
| |
Collapse
|
309
|
Inanc MT, Karadavut S, Aytekin M, Duran AO, Derya M, Akpek M, Sahin O, Kalay N, Karaca H, Ozkan M, Inanc M. The relationship between plasma hyaluronan levels and anthracycline-related cardiotoxicity in breast cancer patients. Int J Cardiol 2016; 218:246-251. [PMID: 27236123 DOI: 10.1016/j.ijcard.2016.05.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 05/12/2016] [Indexed: 11/15/2022]
Abstract
AIM Anthracycline-derived antineoplastic agents are used as the main form of treatment in many malignant diseases, including breast cancer and childhood cancers. Cardiotoxicity is one of the most feared life-threatening complications of cancer therapy. In the present study, we aimed to investigate the relationship between plasma hyaluronan (HA) levels and anthracycline-induced cardiotoxicity. MATERIALS AND METHODS Fifty eight of 73 female patients who were diagnosed with breast cancer and treated with a chemotherapy regimen including anthracycline were enrolled in this study. Anamneses were taken from each patient before and after chemotherapy. Further, physical examinations, electrocardiography, and transthoracic echocardiography were performed, and plasma hyaluronan levels were determined by using ELISA assay for each patient before and after treatment. RESULTS Following anthracycline-based chemotherapy, the average left ventricular ejection fraction decreased (62.6±3.7% vs. 58.6±4.4%, p<0.001), and diastolic functions significantly deteriorated (p<0.001). However, troponin and hyaluronan levels significantly increased following chemotherapy [Troponin (ng/ml, mean±SD): before 0.01±0.002, after 0.037±0.02, p<0.001], [Plasma HA (ng/ml, mean±SD): before 41.3±5.4, after 70±8.5, p<0.001]. The increase in troponin values correlated with systolic dysfunction (p=0.002), but did not correlate with diastolic dysfunction (p=0.661). Significant correlations were found between systolic/diastolic dysfunction and plasma HA levels (r=0.417, p=0.001; r=0.339, p=0.009, respectively). CONCLUSIONS Both systolic and diastolic functions were significantly deteriorated after chemotherapy. In addition, plasma levels of HA and troponin increased after treatment. Further, both systolic and diastolic dysfunctions were found to correlate with serum HA levels. All these data suggest that HA might have a function on anthracycline-induced cardiotoxicity.
Collapse
Affiliation(s)
| | | | - Metin Aytekin
- Erciyes University Medical Biology Department, Kayseri, Turkey
| | - Ayse Ocak Duran
- Erciyes University Medical Oncology Department, Kayseri, Turkey
| | - Muazzez Derya
- Erciyes University Medical Biology Department, Kayseri, Turkey
| | - Mahmut Akpek
- Kayseri Training and Research Hospital Cardiology Department, Kayseri, Turkey
| | - Omer Sahin
- Kayseri Training and Research Hospital Cardiology Department, Kayseri, Turkey
| | - Nihat Kalay
- Erciyes University Cardiology Department, Kayseri, Turkey
| | - Halit Karaca
- Erciyes University Medical Oncology Department, Kayseri, Turkey
| | - Metin Ozkan
- Erciyes University Medical Oncology Department, Kayseri, Turkey
| | - Mevlude Inanc
- Kayseri Training and Research Hospital Medical Oncology Department, Kayseri, Turkey.
| |
Collapse
|
310
|
Das S, Filippone SM, Williams DS, Das A, Kukreja RC. Beet root juice protects against doxorubicin toxicity in cardiomyocytes while enhancing apoptosis in breast cancer cells. Mol Cell Biochem 2016; 421:89-101. [DOI: 10.1007/s11010-016-2789-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 08/05/2016] [Indexed: 12/21/2022]
|
311
|
Schuler MK, Gerdes S, West A, Richter S, Busemann C, Hentschel L, Lenz F, Kopp HG, Ehninger G, Reichardt P, Pink D. Efficacy and safety of Dexrazoxane (DRZ) in sarcoma patients receiving high cumulative doses of anthracycline therapy - a retrospective study including 32 patients. BMC Cancer 2016; 16:619. [PMID: 27507014 PMCID: PMC4977890 DOI: 10.1186/s12885-016-2654-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 07/29/2016] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Anthracyclines, as the most effective therapy, are the cornerstone of advanced stage sarcoma treatment. However, anthracyclines can also contribute to myocardial dysfunction and congestive heart failure, ultimately limiting the therapeutic potential of the drug. Coadministration of Dexrazoxane has been shown to effectively reduce cardiotoxicity, however primarily in patients suffering in diseases other than sarcoma. METHODS The aim of this retrospective analysis was to evaluate safety and efficacy of chemotherapy with high cumulative doses of anthracyclines in combination with Dexrazoxane. The medical charts of 32 patients treated in four institutions were analyzed. Reasons for coadministration were rechallenge, reaching the cumulative anthracycline dose and preexisting heart failure. RESULTS The median age was 54 years [18-68 years]. The median cumulative anthracycline dose before adding DRZ was 450 mg/m(2) and after administration of last anthracycline containing therapy 750 mg/m(2). Either during treatment or follow up, 2/27 patients (7 %) without preexisting major cardiac findings developed anthracycline-induced cardiotoxicity. The median overall survival (OS) from start of the first anthracycline containing chemotherapy was 46 months and 17 months from the initial coadministration of DRZ. At rechallenge, the median progression free survival (PFS) with DRZ was 7 months. In continuous therapy, the median PFS was 13 months from beginning of chemotherapy and 9 months from the addition of DRZ. CONCLUSION Chemotherapy with high cumulative doses of anthracyclines in addition with DRZ demonstrated a remarkable OS in these advanced disease patients. Cardiac side-effects due to high cumulative doses of anthracyclines requiring discontinuation of anthracycline treatment were rare. A PFS of 9 months from the beginning of the coadministration of DRZ indicates that continuing anthracycline therapy beyond established cumulative doses is a promising therapeutic option.
Collapse
Affiliation(s)
- Markus K Schuler
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Technical University Dresden, Fetscherstraße 74, 01307, Dresden, Germany. .,Department of Internal Medicine II, HELIOS Clinic Emil von Behring, Walterhöferstr. 11, 14165, Berlin, Germany.
| | - Sebastian Gerdes
- Institute of Medical Informatics and Biometry, University Hospital Carl Gustav Carus, Technical University Dresden, Fetscherstraße 74, 01307, Dresden, Germany
| | - Antje West
- Department of Hematology and Oncology, HELIOS Clinic Bad Saarow, Pieskower Straße 33, 15526, Bad Saarow, Germany
| | - Stephan Richter
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Technical University Dresden, Fetscherstraße 74, 01307, Dresden, Germany
| | - Christoph Busemann
- Department of Internal Medicine C, University Hospital Greifswald, Sauerbruchstraße, 17475, Greifswald, Germany
| | - Leopold Hentschel
- University Cancer Center, University Hospital Carl Gustav Carus, Technical University Dresden, Fetscherstraße 74, 01307, Dresden, Germany
| | - Felicitas Lenz
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Technical University Dresden, Fetscherstraße 74, 01307, Dresden, Germany
| | - Hans-Georg Kopp
- Department of Internal Medicine II, University Hospital Tübingen, Eberhard Karls University Tübingen, Geissweg 3, 72076, Tübingen, Germany
| | - Gerhard Ehninger
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Technical University Dresden, Fetscherstraße 74, 01307, Dresden, Germany.,University Cancer Center, University Hospital Carl Gustav Carus, Technical University Dresden, Fetscherstraße 74, 01307, Dresden, Germany
| | - Peter Reichardt
- Department of Interdisciplinary Oncology, HELIOS Clinic Berlin-Buch, Berlin, Germany
| | - Daniel Pink
- Department of Hematology and Oncology, HELIOS Clinic Bad Saarow, Pieskower Straße 33, 15526, Bad Saarow, Germany.,Department of Internal Medicine C, University Hospital Greifswald, Sauerbruchstraße, 17475, Greifswald, Germany
| |
Collapse
|
312
|
Buondonno I, Gazzano E, Jean SR, Audrito V, Kopecka J, Fanelli M, Salaroglio IC, Costamagna C, Roato I, Mungo E, Hattinger CM, Deaglio S, Kelley SO, Serra M, Riganti C. Mitochondria-Targeted Doxorubicin: A New Therapeutic Strategy against Doxorubicin-Resistant Osteosarcoma. Mol Cancer Ther 2016; 15:2640-2652. [PMID: 27466354 DOI: 10.1158/1535-7163.mct-16-0048] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 06/23/2016] [Indexed: 11/16/2022]
Abstract
Doxorubicin is one of the leading drugs for osteosarcoma standard chemotherapy. A total of 40% to 45% of high-grade osteosarcoma patients are unresponsive, or only partially responsive, to doxorubicin (Dox), due to the overexpression of the drug efflux transporter ABCB1/P-glycoprotein (Pgp). The aim of this work is to improve Dox-based regimens in resistant osteosarcomas. We used a chemically modified mitochondria-targeted Dox (mtDox) against Pgp-overexpressing osteosarcomas with increased resistance to Dox. Unlike Dox, mtDox accumulated at significant levels intracellularly, exerted cytotoxic activity, and induced necrotic and immunogenic cell death in Dox-resistant/Pgp-overexpressing cells, fully reproducing the activities exerted by anthracyclines in drug-sensitive tumors. mtDox reduced tumor growth and cell proliferation, increased apoptosis, primed tumor cells for recognition by the host immune system, and was less cardiotoxic than Dox in preclinical models of drug-resistant osteosarcoma. The increase in Dox resistance was paralleled by a progressive upregulation of mitochondrial metabolism. By widely modulating the expression of mitochondria-related genes, mtDox decreased mitochondrial biogenesis, the import of proteins and metabolites within mitochondria, mitochondrial metabolism, and the synthesis of ATP. These events were paralleled by increased reactive oxygen species production, mitochondrial depolarization, and mitochondria-dependent apoptosis in resistant osteosarcoma cells, where Dox was completely ineffective. We propose mtDox as a new effective agent with a safer toxicity profile compared with Dox that may be effective for the treatment of Dox-resistant/Pgp-positive osteosarcoma patients, who strongly need alternative and innovative treatment strategies. Mol Cancer Ther; 15(11); 2640-52. ©2016 AACR.
Collapse
Affiliation(s)
| | - Elena Gazzano
- Department of Oncology, University of Torino, Torino, Italy
| | - Sae Rin Jean
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada.,Department of Chemistry, Faculty of Arts and Science, University of Toronto, Toronto, Ontario, Canada
| | - Valentina Audrito
- Human Genetics Foundation (HuGeF), Torino, Italy.,Department of Medical Sciences, University of Torino, Torino, Italy
| | - Joanna Kopecka
- Department of Oncology, University of Torino, Torino, Italy
| | - Marilù Fanelli
- Orthopaedic Rizzoli Institute, Laboratory of Experimental Oncology, Pharmacogenomics and Pharmacogenetics Research Unit, Bologna, Italy
| | | | | | - Ilaria Roato
- Center for Research and Experimental Medicine (Ce.R.M.S.), San Giovanni Battista Hospital, Torino, Italy
| | - Eleonora Mungo
- Department of Oncology, University of Torino, Torino, Italy
| | - Claudia M Hattinger
- Orthopaedic Rizzoli Institute, Laboratory of Experimental Oncology, Pharmacogenomics and Pharmacogenetics Research Unit, Bologna, Italy
| | - Silvia Deaglio
- Human Genetics Foundation (HuGeF), Torino, Italy.,Department of Medical Sciences, University of Torino, Torino, Italy
| | - Shana O Kelley
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada.,Department of Chemistry, Faculty of Arts and Science, University of Toronto, Toronto, Ontario, Canada
| | - Massimo Serra
- Orthopaedic Rizzoli Institute, Laboratory of Experimental Oncology, Pharmacogenomics and Pharmacogenetics Research Unit, Bologna, Italy
| | - Chiara Riganti
- Department of Oncology, University of Torino, Torino, Italy.
| |
Collapse
|
313
|
Liu Y, Li Q, Zhou L, Xie N, Nice EC, Zhang H, Huang C, Lei Y. Cancer drug resistance: redox resetting renders a way. Oncotarget 2016; 7:42740-42761. [PMID: 27057637 PMCID: PMC5173169 DOI: 10.18632/oncotarget.8600] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 03/28/2016] [Indexed: 02/05/2023] Open
Abstract
Disruption of redox homeostasis is a crucial factor in the development of drug resistance, which is a major problem facing current cancer treatment. Compared with normal cells, tumor cells generally exhibit higher levels of reactive oxygen species (ROS), which can promote tumor progression and development. Upon drug treatment, some tumor cells can undergo a process of 'Redox Resetting' to acquire a new redox balance with higher levels of ROS accumulation and stronger antioxidant systems. Evidence has accumulated showing that the 'Redox Resetting' enables cancer cells to become resistant to anticancer drugs by multiple mechanisms, including increased rates of drug efflux, altered drug metabolism and drug targets, activated prosurvival pathways and inefficient induction of cell death. In this article, we provide insight into the role of 'Redox Resetting' on the emergence of drug resistance that may contribute to pharmacological modulation of resistance.
Collapse
Affiliation(s)
- Yuan Liu
- State Key Laboratory for Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, P. R. China
- Department of Neurology, The Affiliated Hospital of Hainan Medical College, Haikou, Hainan, P. R. China
| | - Qifu Li
- Department of Neurology, The Affiliated Hospital of Hainan Medical College, Haikou, Hainan, P. R. China
| | - Li Zhou
- State Key Laboratory for Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, P. R. China
| | - Na Xie
- State Key Laboratory for Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, P. R. China
| | - Edouard C. Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Haiyuan Zhang
- Department of Neurology, The Affiliated Hospital of Hainan Medical College, Haikou, Hainan, P. R. China
| | - Canhua Huang
- State Key Laboratory for Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, P. R. China
| | - Yunlong Lei
- Department of Biochemistry and Molecular Biology, and Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, P. R. China
| |
Collapse
|
314
|
Pathways of cardiac toxicity: comparison between chemotherapeutic drugs doxorubicin and mitoxantrone. Arch Toxicol 2016; 90:2063-2076. [PMID: 27342245 DOI: 10.1007/s00204-016-1759-y] [Citation(s) in RCA: 165] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 06/13/2016] [Indexed: 01/25/2023]
Abstract
Anthracyclines, e.g., doxorubicin (DOX), and anthracenediones, e.g., mitoxantrone (MTX), are drugs used in the chemotherapy of several cancer types, including solid and non-solid malignancies such as breast cancer, leukemia, lymphomas, and sarcomas. Although they are effective in tumor therapy, treatment with these two drugs may lead to side effects such as arrhythmia and heart failure. At the same clinically equivalent dose, MTX causes slightly reduced cardiotoxicity compared with DOX. These drugs interact with iron to generate reactive oxygen species (ROS), target topoisomerase 2 (Top2), and impair mitochondria. These are some of the mechanisms through which these drugs induce late cardiomyopathy. In this review, we compare the cardiotoxicities of these two chemotherapeutic drugs, DOX and MTX. As described here, even though they share similarities in their modes of toxicant action, DOX and MTX seem to differ in a key aspect. DOX is a more redox-interfering drug, while MTX induces energy imbalance. In addition, DOX toxicity can be explained by underlying mechanisms that include targeting of Top2 beta, mitochondrial impairment, and increases in ROS generation. These modes of action have not yet been demonstrated for MTX, and this knowledge gap needs to be filled.
Collapse
|
315
|
Sági JC, Kutszegi N, Kelemen A, Fodor LE, Gézsi A, Kovács GT, Erdélyi DJ, Szalai C, Semsei ÁF. Pharmacogenetics of anthracyclines. Pharmacogenomics 2016; 17:1075-87. [DOI: 10.2217/pgs-2016-0036] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Anthracyclines constitute a fundamental part of the chemotherapy regimens utilized to treat a number of different malignancies both in pediatric and adult patients. These drugs are one of the most efficacious anticancer agents ever invented. On the other hand, anthracyclines are cardiotoxic. Childhood cancer survivors treated with anthracyclines often undergo cardiac complications which are influenced by genetic variations of the patients. The scientific literature comprises numerous investigations in the subject of the pharmacogenetics of anthracyclines. In this review, we provide a comprehensive overview of this research topic. Genetic variants are proposed targets in the personalized treatment in order to individualize dosing and therefore reduce side effects.
Collapse
Affiliation(s)
- Judit C Sági
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, H-1089 Budapest, Nagyvárad tér 4, Hungary
| | - Nóra Kutszegi
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, H-1089 Budapest, Nagyvárad tér 4, Hungary
| | - Andrea Kelemen
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, H-1089 Budapest, Nagyvárad tér 4, Hungary
| | - Lili E Fodor
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, H-1089 Budapest, Nagyvárad tér 4, Hungary
| | - András Gézsi
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, H-1089 Budapest, Nagyvárad tér 4, Hungary
| | - Gábor T Kovács
- Second Department of Pediatrics, Semmelweis University, H-1094 Budapest, Tűzoltó utca 7–9, Hungary
| | - Dániel J Erdélyi
- Second Department of Pediatrics, Semmelweis University, H-1094 Budapest, Tűzoltó utca 7–9, Hungary
| | - Csaba Szalai
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, H-1089 Budapest, Nagyvárad tér 4, Hungary
- Central Laboratory, Heim Pal Children Hospital, H-1089 Budapest, Üllői út 86, Hungary
| | - Ágnes F Semsei
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, H-1089 Budapest, Nagyvárad tér 4, Hungary
| |
Collapse
|
316
|
Cardioprotective Potentials of Plant-Derived Small Molecules against Doxorubicin Associated Cardiotoxicity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:5724973. [PMID: 27313831 PMCID: PMC4893565 DOI: 10.1155/2016/5724973] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 04/02/2016] [Accepted: 04/20/2016] [Indexed: 12/17/2022]
Abstract
Doxorubicin (DOX) is a potent and widely used anthracycline antibiotic for the treatment of several malignancies. Unfortunately, the clinical utility of DOX is often restricted due to the elicitation of organ toxicity. Particularly, the increased risk for the development of dilated cardiomyopathy by DOX among the cancer survivors warrants major attention from the physicians as well as researchers to develop adjuvant agents to neutralize the noxious effects of DOX on the healthy myocardium. Despite these pitfalls, the use of traditional cytotoxic drugs continues to be the mainstay treatment for several types of cancer. Recently, phytochemicals have gained attention for their anticancer, chemopreventive, and cardioprotective activities. The ideal cardioprotective agents should not compromise the clinical efficacy of DOX and should be devoid of cumulative or irreversible toxicity on the naïve tissues. Furthermore, adjuvants possessing synergistic anticancer activity and quelling of chemoresistance would significantly enhance the clinical utility in combating DOX-induced cardiotoxicity. The present review renders an overview of cardioprotective effects of plant-derived small molecules and their purported mechanisms against DOX-induced cardiotoxicity. Phytochemicals serve as the reservoirs of pharmacophore which can be utilized as templates for developing safe and potential novel cardioprotective agents in combating DOX-induced cardiotoxicity.
Collapse
|
317
|
Srankova J, Doka G, Pivackova L, Mesarosova L, Kyselovic J, Klimas J, Krenek P. Daunorubicin Down-Regulates the Expression of Stem Cell Markers and Factors Involved in Stem Cell Migration and Homing in Rat Heart in Subchronic but not Acute Cardiomyopathy. Basic Clin Pharmacol Toxicol 2016; 119:443-452. [PMID: 27090888 DOI: 10.1111/bcpt.12606] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 04/06/2016] [Indexed: 12/26/2022]
Abstract
We tested the hypothesis that daunorubicin (DAU) cardiotoxicity alters expression of cytokines involved in stem cell migration and homing. Male Wistar rats were treated with daunorubicin to induce acute DAU cardiomyopathy (6 × 3 mg/kg, i.p., every 48 hr, DAU-A) or subchronic DAU cardiomyopathy (15 mg/kg, i.v., DAU-C). The left ventricle was catheterized. The animals were killed 48 hr (DAU-A) and 8 weeks (DAU-C) after the last dose of DAU. Expression of foetal genes (Nppa, Nppb), isomyosins (Myh6, Myh7), sources of oxidative stress (Abcb8, gp91phox), cytokines (Sdf-1, Cxcr4, Scf, Vegf, Hgf, Igf-1), markers of cardiac progenitor (c-kit, Atnx-1), endothelial progenitor (CD34, CD133) and mesenchymal (CD44, CD105) stem cells were determined by qRT-PCR in left ventricular tissue. Reduced body-weight, decreased left ventricular weight and function, and elevated Nppa, Nppb, Myh7 were observed in both models. Myh6 decreased only in DAU-C, which had a 35% mortality. Up-regulated gp91phox and down-regulated Abcb8 in DAU were present only in DAU-C where we observed markedly decreased expressions of Scf and Vegf as well as expressions of stem cell markers. Down-regulation of cytokines and stem cell markers may reflect impaired chemotaxis, migration and homing of stem cells and tissue repair in the heart in subchronic but not acute model of DAU cardiomyopathy.
Collapse
Affiliation(s)
- Jasna Srankova
- Comenius University in Bratislava, Faculty of Pharmacy, Department of Pharmacology and Toxicology, Bratislava, Slovak Republic
| | - Gabriel Doka
- Comenius University in Bratislava, Faculty of Pharmacy, Department of Pharmacology and Toxicology, Bratislava, Slovak Republic
| | - Lenka Pivackova
- Comenius University in Bratislava, Faculty of Pharmacy, Department of Pharmacology and Toxicology, Bratislava, Slovak Republic
| | - Lucia Mesarosova
- Comenius University in Bratislava, Faculty of Pharmacy, Department of Pharmacology and Toxicology, Bratislava, Slovak Republic
| | - Jan Kyselovic
- Comenius University in Bratislava, Faculty of Pharmacy, Department of Pharmacology and Toxicology, Bratislava, Slovak Republic
| | - Jan Klimas
- Comenius University in Bratislava, Faculty of Pharmacy, Department of Pharmacology and Toxicology, Bratislava, Slovak Republic
| | - Peter Krenek
- Comenius University in Bratislava, Faculty of Pharmacy, Department of Pharmacology and Toxicology, Bratislava, Slovak Republic.
| |
Collapse
|
318
|
Chegaev K, Rolando B, Cortese D, Gazzano E, Buondonno I, Lazzarato L, Fanelli M, Hattinger CM, Serra M, Riganti C, Fruttero R, Ghigo D, Gasco A. H2S-Donating Doxorubicins May Overcome Cardiotoxicity and Multidrug Resistance. J Med Chem 2016; 59:4881-9. [PMID: 27120394 DOI: 10.1021/acs.jmedchem.6b00184] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Doxorubicin (DOXO) is one of the most effective antineoplastic agents in clinical practice. Its use is limited by acute and chronic side effects, in particular by its cardiotoxicity and by the rapid development of resistance to it. As part of a program aimed at developing new DOXO derivatives endowed with reduced cardiotoxicity, and active against DOXO-resistant tumor cells, a series of H2S-releasing DOXOs (H2S-DOXOs) were obtained by combining DOXO with appropriate H2S donor substructures. The resulting compounds were studied on H9c2 cardiomyocytes and in DOXO-sensitive U-2OS osteosarcoma cells, as well as in related cell variants with increasing degrees of DOXO-resistance. Differently from DOXO, most of the products were not toxic at 5 μM concentration on H9c2 cells. A few of them triggered high activity on the cancer cells. H2S-DOXOs 10 and 11 emerged as the most interesting members of the series. The capacity of 10 to impair Pgp transporter is also discussed.
Collapse
Affiliation(s)
- Konstantin Chegaev
- Department of Science and Drug Technology, University of Torino , via Pietro Giuria 9, 10125 Torino, Italy
| | - Barbara Rolando
- Department of Science and Drug Technology, University of Torino , via Pietro Giuria 9, 10125 Torino, Italy
| | - Daniela Cortese
- Department of Science and Drug Technology, University of Torino , via Pietro Giuria 9, 10125 Torino, Italy
| | - Elena Gazzano
- Department of Oncology and Center for Experimental Research and Medical Studies (CeRMS), University of Torino , via Santena, 5/bis, 10126 Torino, Italy
| | - Ilaria Buondonno
- Department of Oncology and Center for Experimental Research and Medical Studies (CeRMS), University of Torino , via Santena, 5/bis, 10126 Torino, Italy
| | - Loretta Lazzarato
- Department of Science and Drug Technology, University of Torino , via Pietro Giuria 9, 10125 Torino, Italy
| | - Marilù Fanelli
- Orthopaedic Rizzoli Institute, Laboratory of Experimental Oncology, Pharmacogenomics and Pharmacogenetics Research Unit , via G. C. Pupilli 1, 40136 Bologna, Italy
| | - Claudia M Hattinger
- Orthopaedic Rizzoli Institute, Laboratory of Experimental Oncology, Pharmacogenomics and Pharmacogenetics Research Unit , via G. C. Pupilli 1, 40136 Bologna, Italy
| | - Massimo Serra
- Orthopaedic Rizzoli Institute, Laboratory of Experimental Oncology, Pharmacogenomics and Pharmacogenetics Research Unit , via G. C. Pupilli 1, 40136 Bologna, Italy
| | - Chiara Riganti
- Department of Oncology and Center for Experimental Research and Medical Studies (CeRMS), University of Torino , via Santena, 5/bis, 10126 Torino, Italy
| | - Roberta Fruttero
- Department of Science and Drug Technology, University of Torino , via Pietro Giuria 9, 10125 Torino, Italy
| | - Dario Ghigo
- Department of Oncology and Center for Experimental Research and Medical Studies (CeRMS), University of Torino , via Santena, 5/bis, 10126 Torino, Italy
| | - Alberto Gasco
- Department of Science and Drug Technology, University of Torino , via Pietro Giuria 9, 10125 Torino, Italy
| |
Collapse
|
319
|
Maillet A, Tan K, Chai X, Sadananda SN, Mehta A, Ooi J, Hayden MR, Pouladi MA, Ghosh S, Shim W, Brunham LR. Modeling Doxorubicin-Induced Cardiotoxicity in Human Pluripotent Stem Cell Derived-Cardiomyocytes. Sci Rep 2016; 6:25333. [PMID: 27142468 PMCID: PMC4855185 DOI: 10.1038/srep25333] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 04/15/2016] [Indexed: 12/12/2022] Open
Abstract
Doxorubicin is a highly efficacious anti-cancer drug but causes cardiotoxicity in many patients. The mechanisms of doxorubicin-induced cardiotoxicity (DIC) remain incompletely understood. We investigated the characteristics and molecular mechanisms of DIC in human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs). We found that doxorubicin causes dose-dependent increases in apoptotic and necrotic cell death, reactive oxygen species production, mitochondrial dysfunction and increased intracellular calcium concentration. We characterized genome-wide changes in gene expression caused by doxorubicin using RNA-seq, as well as electrophysiological abnormalities caused by doxorubicin with multi-electrode array technology. Finally, we show that CRISPR-Cas9-mediated disruption of TOP2B, a gene implicated in DIC in mouse studies, significantly reduces the sensitivity of hPSC-CMs to doxorubicin-induced double stranded DNA breaks and cell death. These data establish a human cellular model of DIC that recapitulates many of the cardinal features of this adverse drug reaction and could enable screening for protective agents against DIC as well as assessment of genetic variants involved in doxorubicin response.
Collapse
Affiliation(s)
- Agnes Maillet
- Translational Laboratory in Genetic Medicine, National University of Singapore and the Agency for Science Technology and Research (A*STAR), Singapore
| | - Kim Tan
- Translational Laboratory in Genetic Medicine, National University of Singapore and the Agency for Science Technology and Research (A*STAR), Singapore
| | - Xiaoran Chai
- Center for Computational Biology, Duke-NUS Graduate Medical School, Singapore
| | - Singh N Sadananda
- Translational Laboratory in Genetic Medicine, National University of Singapore and the Agency for Science Technology and Research (A*STAR), Singapore
| | - Ashish Mehta
- National Heart Research Institute, National Heart Centre Singapore, Singapore.,Cardiovascular Academic Clinical Program, DUKE-NUS Graduate Medical School, Singapore
| | - Jolene Ooi
- Translational Laboratory in Genetic Medicine, National University of Singapore and the Agency for Science Technology and Research (A*STAR), Singapore
| | - Michael R Hayden
- Translational Laboratory in Genetic Medicine, National University of Singapore and the Agency for Science Technology and Research (A*STAR), Singapore.,Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, Canada
| | - Mahmoud A Pouladi
- Translational Laboratory in Genetic Medicine, National University of Singapore and the Agency for Science Technology and Research (A*STAR), Singapore.,Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Sujoy Ghosh
- Center for Computational Biology, Duke-NUS Graduate Medical School, Singapore.,Program in Cardiovascular and Metabolic Disorders, Duke-NUS Graduate Medical School, Singapore
| | - Winston Shim
- National Heart Research Institute, National Heart Centre Singapore, Singapore.,Cardiovascular Academic Clinical Program, DUKE-NUS Graduate Medical School, Singapore
| | - Liam R Brunham
- Translational Laboratory in Genetic Medicine, National University of Singapore and the Agency for Science Technology and Research (A*STAR), Singapore.,Department of Medicine, Centre for Heart Lung Innovation, University of British Columbia, Vancouver, Canada
| |
Collapse
|
320
|
van Gisbergen MW, Cebula M, Zhang J, Ottosson-Wadlund A, Dubois L, Lambin P, Tew KD, Townsend DM, Haenen GRMM, Drittij-Reijnders MJ, Saneyoshi H, Araki M, Shishido Y, Ito Y, Arnér ESJ, Abe H, Morgenstern R, Johansson K. Chemical Reactivity Window Determines Prodrug Efficiency toward Glutathione Transferase Overexpressing Cancer Cells. Mol Pharm 2016; 13:2010-25. [PMID: 27093577 DOI: 10.1021/acs.molpharmaceut.6b00140] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Glutathione transferases (GSTs) are often overexpressed in tumors and frequently correlated to bad prognosis and resistance against a number of different anticancer drugs. To selectively target these cells and to overcome this resistance we previously have developed prodrugs that are derivatives of existing anticancer drugs (e.g., doxorubicin) incorporating a sulfonamide moiety. When cleaved by GSTs, the prodrug releases the cytostatic moiety predominantly in GST overexpressing cells, thus sparing normal cells with moderate enzyme levels. By modifying the sulfonamide it is possible to control the rate of drug release and specifically target different GSTs. Here we show that the newly synthesized compounds, 4-acetyl-2-nitro-benzenesulfonyl etoposide (ANS-etoposide) and 4-acetyl-2-nitro-benzenesulfonyl doxorubicin (ANS-DOX), function as prodrugs for GSTA1 and MGST1 overexpressing cell lines. ANS-DOX, in particular, showed a desirable cytotoxic profile by inducing toxicity and DNA damage in a GST-dependent manner compared to control cells. Its moderate conversion of 500 nmol/min/mg, as catalyzed by GSTA1, seems hereby essential since the more reactive 2,4-dinitrobenzenesulfonyl doxorubicin (DNS-DOX) (14000 nmol/min/mg) did not display a preference for GSTA1 overexpressing cells. DNS-DOX, however, effectively killed GSTP1 (20 nmol/min/mg) and MGST1 (450 nmol/min/mg) overexpressing cells as did the less reactive 4-mononitrobenzenesulfonyl doxorubicin (MNS-DOX) in a MGST1-dependent manner (1.5 nmol/min/mg) as shown previously. Furthermore, we show that the mechanism of these prodrugs involves a reduction in GSH levels as well as inhibition of the redox regulatory enzyme thioredoxin reductase 1 (TrxR1) by virtue of their electrophilic sulfonamide moiety. TrxR1 is upregulated in many tumors and associated with resistance to chemotherapy and poor patient prognosis. Additionally, the prodrugs potentially acted as a general shuttle system for DOX, by overcoming resistance mechanisms in cells. Here we propose that GST-dependent prodrugs require a conversion rate "window" in order to selectively target GST overexpressing cells, while limiting their effects on normal cells. Prodrugs are furthermore a suitable system to specifically target GSTs and to overcome various drug resistance mechanisms that apply to the parental drug.
Collapse
Affiliation(s)
- Marike W van Gisbergen
- Institute of Environmental Medicine, Division of Biochemical Toxicology, Karolinska Institutet , SE 17177 Stockholm, Sweden.,Department of Radiation Oncology (MaastRO Lab), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center , Universiteitssingel 50/23, PO Box 616, 6200 MD Maastricht, The Netherlands
| | - Marcus Cebula
- Institute of Environmental Medicine, Division of Biochemical Toxicology, Karolinska Institutet , SE 17177 Stockholm, Sweden
| | - Jie Zhang
- Institute of Environmental Medicine, Division of Biochemical Toxicology, Karolinska Institutet , SE 17177 Stockholm, Sweden.,Departments of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina , Charleston, South Carolina 29425, United States
| | - Astrid Ottosson-Wadlund
- Institute of Environmental Medicine, Division of Biochemical Toxicology, Karolinska Institutet , SE 17177 Stockholm, Sweden
| | - Ludwig Dubois
- Department of Radiation Oncology (MaastRO Lab), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center , Universiteitssingel 50/23, PO Box 616, 6200 MD Maastricht, The Netherlands
| | - Philippe Lambin
- Department of Radiation Oncology (MaastRO Lab), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center , Universiteitssingel 50/23, PO Box 616, 6200 MD Maastricht, The Netherlands
| | - Kenneth D Tew
- Departments of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina , Charleston, South Carolina 29425, United States
| | - Danyelle M Townsend
- Department of Pharmaceutical and Biomedical Sciences, Medical University of South Carolina , Charleston, South Carolina 29425, United States
| | - Guido R M M Haenen
- Department of Toxicology, NUTRIM-School for Nutrition, Toxicology, and Metabolism, Maastricht University Medical Center , Universiteitssingel 50/23, PO Box 616, 6200 MD Maastricht, The Netherlands
| | - Marie-José Drittij-Reijnders
- Department of Toxicology, NUTRIM-School for Nutrition, Toxicology, and Metabolism, Maastricht University Medical Center , Universiteitssingel 50/23, PO Box 616, 6200 MD Maastricht, The Netherlands
| | - Hisao Saneyoshi
- Nano Medical Engineering Laboratory, Discovery Research Institute, RIKEN 2-1 , Hirosawa, Wako-Shi, Saitama 351-0198, Japan
| | - Mika Araki
- Nano Medical Engineering Laboratory, Discovery Research Institute, RIKEN 2-1 , Hirosawa, Wako-Shi, Saitama 351-0198, Japan
| | - Yuko Shishido
- Department of Chemistry, Graduate School of Science, Nagoya University , Furo-cho, Chikusa-ku, Nagoya 464-8602, Japan
| | - Yoshihiro Ito
- Nano Medical Engineering Laboratory, Discovery Research Institute, RIKEN 2-1 , Hirosawa, Wako-Shi, Saitama 351-0198, Japan
| | - Elias S J Arnér
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet , SE-171 77 Stockholm, Sweden
| | - Hiroshi Abe
- Nano Medical Engineering Laboratory, Discovery Research Institute, RIKEN 2-1 , Hirosawa, Wako-Shi, Saitama 351-0198, Japan.,Department of Chemistry, Graduate School of Science, Nagoya University , Furo-cho, Chikusa-ku, Nagoya 464-8602, Japan
| | - Ralf Morgenstern
- Institute of Environmental Medicine, Division of Biochemical Toxicology, Karolinska Institutet , SE 17177 Stockholm, Sweden
| | - Katarina Johansson
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet , SE-171 77 Stockholm, Sweden
| |
Collapse
|
321
|
Xi L. Visnagin-a new protectant against doxorubicin cardiotoxicity? Inhibition of mitochondrial malate dehydrogenase 2 (MDH2) and beyond. ANNALS OF TRANSLATIONAL MEDICINE 2016; 4:65. [PMID: 27004212 DOI: 10.3978/j.issn.2305-5839.2015.10.43] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Lei Xi
- Pauley Heart Center, Division of Cardiology, Virginia Commonwealth University, Richmond, VA 23298-0204, USA
| |
Collapse
|
322
|
Ren WX, Han J, Uhm S, Jang YJ, Kang C, Kim JH, Kim JS. Recent development of biotin conjugation in biological imaging, sensing, and target delivery. Chem Commun (Camb) 2016; 51:10403-18. [PMID: 26021457 DOI: 10.1039/c5cc03075g] [Citation(s) in RCA: 264] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Despite encouraging results from preliminary studies of anticancer therapies, the lack of tumor specificity remains an important issue in the modern pharmaceutical industry. New findings indicate that biotin or biotin-conjugates could be favorably assimilated by tumor cells that over-express biotin-selective transporters. Furthermore, biotin can form stable complexes with avidin and its bacterial counterpart streptavidin. The strong bridging between avidin and biotin moieties on other molecules is a proven adaptable tool with broad biological applications. Under these circumstances, a biotin moiety is certainly an attractive choice for live-cell imaging, biosensing, and target delivery.
Collapse
Affiliation(s)
- Wen Xiu Ren
- Department of Chemistry, Korea University, Seoul 136-701, South Korea.
| | | | | | | | | | | | | |
Collapse
|
323
|
Abstract
Long term survival of childhood cancers is now more than 70%. Anthracyclines, including doxorubicin, are some of the most efficacious anticancer drugs available. However, its use as a chemotherapeutic agent is severely hindered by its dose-limiting toxicities. Most notably observed is cardiotoxicity, but other organ systems are also degraded by doxorubicin use. Despite the years of its use and the amount of information written about this drug, an understanding of its cellular mechanisms is not fully appreciated. The mechanisms by which doxorubicin induces cytotoxicity in target cancer cells have given insight about how the drug damages cardiomyocytes. The major mechanisms of doxorubicin actions are thought to be as an oxidant generator and as an inhibitor of topoisomerase 2. However, other signaling pathways are also invoked with significant consequences for the cardiomyocyte. Further the interaction between oxidant generation and topoisomerase function has only recently been appreciated and the consequences of this interaction are still not fully understood. The unfortunate consequences of doxorubicin within cardiomyocytes have promoted the search for new drugs and methods that can prevent or reverse the damage caused to the heart after treatment in cancer patients. Alternative protocols have lessened the impact on newly diagnosed cancer patients. However the years of doxorubicin use have generated a need for monitoring the onset of cardiotoxicity as well as understanding its potential long-term consequences. Although a fairly clear understanding of the short-term pathologic mechanisms of doxorubicin actions has been achieved, the long-term mechanisms of doxorubicin induced heart failure remain to be carefully delineated.
Collapse
Affiliation(s)
| | - John G. Edwards
- Corresponding author at: Department of Physiology, New York Medical College, 15 Dana Road, Valhalla, NY, United States.Department of PhysiologyNew York Medical College15 Dana RoadValhallaNYUnited States
| |
Collapse
|
324
|
Yu AF, Jones LW. Breast cancer treatment-associated cardiovascular toxicity and effects of exercise countermeasures. CARDIO-ONCOLOGY 2016; 2:1. [PMID: 28133540 PMCID: PMC5268817 DOI: 10.1186/s40959-016-0011-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Advances in breast cancer treatment have improved disease-free survival and overall survival in women with early-stage breast cancer. However, these improvements may be attenuated by the adverse cardiovascular effects associated with breast cancer adjuvant therapy. Exercise may be a potential strategy to counteract these toxicities. The purpose of this paper is to provide an overview on the adverse cardiovascular effects of breast cancer therapy as well as the evidence supporting the potential cardioprotective effects of exercise training in breast cancer patients during and after treatment. We will also discuss research gaps and avenues for future research.
Collapse
Affiliation(s)
- Anthony F Yu
- Department of Medicine, Memorial Sloan Kettering Cancer Center, Cardiology Service, 1275 York Avenue, New York, NY, 10065, USA
| | - Lee W Jones
- Department of Medicine, Memorial Sloan Kettering Cancer Center, Cardiology Service, 1275 York Avenue, New York, NY, 10065, USA.
| |
Collapse
|
325
|
Redox cycling metals: Pedaling their roles in metabolism and their use in the development of novel therapeutics. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:727-48. [PMID: 26844773 DOI: 10.1016/j.bbamcr.2016.01.026] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 01/29/2016] [Indexed: 12/12/2022]
Abstract
Essential metals, such as iron and copper, play a critical role in a plethora of cellular processes including cell growth and proliferation. However, concomitantly, excess of these metal ions in the body can have deleterious effects due to their ability to generate cytotoxic reactive oxygen species (ROS). Thus, the human body has evolved a very well-orchestrated metabolic system that keeps tight control on the levels of these metal ions. Considering their very high proliferation rate, cancer cells require a high abundance of these metals compared to their normal counterparts. Interestingly, new anti-cancer agents that take advantage of the sensitivity of cancer cells to metal sequestration and their susceptibility to ROS have been developed. These ligands can avidly bind metal ions to form redox active metal complexes, which lead to generation of cytotoxic ROS. Furthermore, these agents also act as potent metastasis suppressors due to their ability to up-regulate the metastasis suppressor gene, N-myc downstream regulated gene 1. This review discusses the importance of iron and copper in the metabolism and progression of cancer, how they can be exploited to target tumors and the clinical translation of novel anti-cancer chemotherapeutics.
Collapse
|
326
|
Wickramasinghe CD, Nguyen KL, Watson KE, Vorobiof G, Yang EH. Concepts in cardio-oncology: definitions, mechanisms, diagnosis and treatment strategies of cancer therapy-induced cardiotoxicity. Future Oncol 2016; 12:855-70. [PMID: 26829050 DOI: 10.2217/fon.15.349] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
There has been considerable improvement in cancer survival rates, primarily through improved preventive strategies and novel anticancer drugs. Cancer is now becoming a chronic illness and as such both short and long-term cardiotoxic effects of cancer therapy are becoming more apparent. This has led to the emergence of a new multidisciplinary specialty known as cardio-oncology, with the purpose of identifying patients who are at a higher risk for developing cardiotoxicity so that appropriate surveillance, treatment and follow-up strategies may be instituted early. The mechanisms of cardiotoxicity caused by commonly used anticancer agents are reviewed, along with the latest advances in diagnostic and preventative strategies, with the overall objective of allowing cancer patients to continue both lifesaving and palliative treatments for their malignancy.
Collapse
Affiliation(s)
- Chanaka D Wickramasinghe
- Division of Cardiology, Department of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| | - Kim-Lien Nguyen
- Division of Cardiology, Department of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| | - Karol E Watson
- Division of Cardiology, Department of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| | - Gabriel Vorobiof
- Division of Cardiology, Department of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| | - Eric H Yang
- Division of Cardiology, Department of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
327
|
New signal transduction paradigms in anthracycline-induced cardiotoxicity. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:1916-25. [PMID: 26828775 DOI: 10.1016/j.bbamcr.2016.01.021] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 01/06/2016] [Accepted: 01/28/2016] [Indexed: 12/21/2022]
Abstract
Anthracyclines, such as doxorubicin, are the most potent and widely used chemotherapeutic agents for the treatment of a variety of human cancers, including solid tumors and hematological malignancies. However, their clinical use is hampered by severe cardiotoxic side effects and cancer therapy-related heart disease has become a leading cause of morbidity and mortality among cancer survivors. The identification of therapeutic strategies limiting anthracycline cardiotoxicity with preserved antitumor efficacy thus represents the current challenge of cardio-oncologists. Anthracycline cardiotoxicity has been originally ascribed to the ability of this class of drugs to disrupt iron metabolism and generate excess of reactive oxygen species (ROS). However, small clinical trials with iron chelators and anti-oxidants failed to provide any benefit and suggested that doxorubicin cardiotoxicity is not solely due to redox cycling. New emerging explanations include anthracycline-dependent regulation of major signaling pathways controlling DNA damage response, cardiomyocyte survival, cardiac inflammation, energetic stress and gene expression modulation. This review will summarize recent studies unraveling the complex web of mechanisms of doxorubicin-mediated cardiotoxicity, and identifying new druggable players for the prevention of heart disease in cancer patients. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Integration of Developmental and Environmental Cues in the Heart edited by Marcus Schaub and Hughes Abriel.
Collapse
|
328
|
The role of oxidative stress on breast cancer development and therapy. Tumour Biol 2016; 37:4281-91. [PMID: 26815507 DOI: 10.1007/s13277-016-4873-9] [Citation(s) in RCA: 157] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 01/15/2016] [Indexed: 02/07/2023] Open
Abstract
Reactive oxygen species (ROS) are produced by both enzymatic and non-enzymatic systems within eukaryotic cells and play important roles in cellular physiology and pathophysiology. Although physiological concentrations are crucial for ensuring cell survival, ROS overproduction is detrimental to cells, and considered key-factors for the development of several diseases, such as neurodegenerative diseases, cardiovascular disorders, and cancer. Cancer cells are usually submitted to higher ROS levels that further stimulate malignant phenotype through stimulus to sustained proliferation, death evasion, angiogenesis, invasiveness, and metastasis. The role of ROS on breast cancer etiology and progression is being progressively elucidated. However, less attention has been given to the development of redox system-targeted strategies for breast cancer therapy. In this review, we address the basic mechanisms of ROS production and scavenging in breast tumor cells, and the emerging possibilities of breast cancer therapies targeting ROS homeostasis.
Collapse
|
329
|
Poręba M, Gać P, Usnarska-Zubkiewicz L, Pilecki W, Kuliczkowski K, Mazur G, Sobieszczańska M, Poręba R. Echocardiographic evaluation of the early cardiotoxic effect of hematopoietic stem cell transplantation in patients with hematologic malignancies. Leuk Lymphoma 2016; 57:2119-25. [PMID: 26762118 DOI: 10.3109/10428194.2015.1122782] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The purpose was to evaluate the early cardiotoxic effects of the treatment in the course of hematopoietic stem cell transplantation (HSCT) in patients with hematologic malignancies. The studies were conducted on 47 patients qualified for the HSCT. Echocardiography was carried out prior to the HSCT and after the HSCT. It was shown that higher age, administration of cyclophosphamide and higher glucose concentrations represented independent risk factors for the worsening of left ventricular diastolic function. Higher cumulative dose of anthracyclines in the previous cytostatic treatment, higher age and administration of cyclophosphamide represented independent risk factors for worsening of left ventricular systolic function. Peri-transplant therapy in the course of HSCT in patients with hematologic malignancies gives the negative effect on the diastolic and systolic left ventricular function, however, previous treatment is of importance, as higher cumulative dose of anthracyclines represents an independent risk factor for the worsening of left ventricular systolic function.
Collapse
Affiliation(s)
- Małgorzata Poręba
- a Department of Pathophysiology , Wroclaw Medical University , Wroclaw , Poland
| | - Paweł Gać
- a Department of Pathophysiology , Wroclaw Medical University , Wroclaw , Poland
| | - Lidia Usnarska-Zubkiewicz
- b Department of Hematology , Blood Neoplasms and Bone Marrow Transplantation, Wroclaw Medical University , Wroclaw , Poland
| | - Witold Pilecki
- a Department of Pathophysiology , Wroclaw Medical University , Wroclaw , Poland
| | - Kazimierz Kuliczkowski
- b Department of Hematology , Blood Neoplasms and Bone Marrow Transplantation, Wroclaw Medical University , Wroclaw , Poland
| | - Grzegorz Mazur
- c Department of Internal Medicine , Occupational Diseases and Hypertension, Wroclaw Medical University , Wroclaw , Poland
| | | | - Rafał Poręba
- c Department of Internal Medicine , Occupational Diseases and Hypertension, Wroclaw Medical University , Wroclaw , Poland
| |
Collapse
|
330
|
Carvalho Rodrigues MA, dos Santos NAG, da Silva Faria MC, Rodrigues JL, Kinoshita A, Baffa O, Antunes LMG, Barbosa F, Gobe GC, dos Santos AC. Carvedilol protects the kidneys of tumor-bearing mice without impairing the biodistribution or the genotoxicity of cisplatin. Chem Biol Interact 2016; 245:59-65. [PMID: 26751708 DOI: 10.1016/j.cbi.2015.12.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 12/23/2015] [Accepted: 12/29/2015] [Indexed: 10/22/2022]
Abstract
Cisplatin (Cisp) is an effective antitumor drug; however, it causes severe nephrotoxicity. Minimization of renal toxicity is essential, but the interference of nephroprotective agents, particularly antioxidants, with the antitumor activity of cisplatin is a general concern. We have recently demonstrated that the anti-hypertensive and antioxidant drug carvedilol (CV) protects against the renal damage and increases the survival of tumor-bearing mice without impairing the tumor reduction by cisplatin. So far, reports on the antioxidant mechanism of CV are controversial and there are no data on the impact of CV on the antitumor mechanisms of cisplatin. Therefore, this study addresses the effect of CV on mechanisms underlying the tumor control by cisplatin. CV did not interfere with the biodistribution or the genotoxicity of cisplatin. We also addressed the antioxidant mechanisms of CV and demonstrated that it does not neutralize free radicals, but is an efficient chelator of ferrous ions that are relevant catalyzers in cisplatin nephrotoxicity. The present data suggest that oxidative damage and genotoxicity play different roles in the toxicity of cisplatin on kidneys and tumors and therefore, some antioxidants might be safe as chemoprotectors. Altogether, our studies provide consistent evidence of the beneficial effect of CV on animals treated with cisplatin and might encourage clinical trials.
Collapse
Affiliation(s)
- Maria A Carvalho Rodrigues
- Centro Universitário do Distrito Federal, UDF, SEP/SUL EQ704/904 Conj.A, Brasilia/DF, CEP 70390-045, Brazil
| | - Neife A G dos Santos
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto-USP, Av. do Café s/n, 14040-903, Ribeirão Preto, SP, Brazil
| | - Marcia C da Silva Faria
- Universidade Federal do Vale do Jequitinhona e Mucuri, UFVJM, Campus do Mucuri, Teófilo Otoni/MG, Rua do Cruzeiro, nº 01 - Jardim, São Paulo, CEP 39803-371, Brazil
| | - Jairo Lisboa Rodrigues
- Universidade Federal do Vale do Jequitinhona e Mucuri, UFVJM, Campus do Mucuri, Teófilo Otoni/MG, Rua do Cruzeiro, nº 01 - Jardim, São Paulo, CEP 39803-371, Brazil
| | - Angela Kinoshita
- Departamento de Física, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto - USP, Avenida do Café s/n, 14040-901, Ribeirão Preto, SP, Brazil
| | - Oswaldo Baffa
- Departamento de Física, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto - USP, Avenida do Café s/n, 14040-901, Ribeirão Preto, SP, Brazil
| | - Lusania M Greggi Antunes
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto-USP, Av. do Café s/n, 14040-903, Ribeirão Preto, SP, Brazil
| | - Fernando Barbosa
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto-USP, Av. do Café s/n, 14040-903, Ribeirão Preto, SP, Brazil
| | - Glenda C Gobe
- Centre for Kidney Disease Research, School of Medicine, The University of Queensland at Princess Alexandra Hospital, Brisbane, QLD, 4102, Australia
| | - Antonio Cardozo dos Santos
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto-USP, Av. do Café s/n, 14040-903, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
331
|
Effect of Metformin and Sitagliptin on Doxorubicin-Induced Cardiotoxicity in Rats: Impact of Oxidative Stress, Inflammation, and Apoptosis. J Toxicol 2015; 2015:424813. [PMID: 26880912 PMCID: PMC4736207 DOI: 10.1155/2015/424813] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 12/14/2015] [Indexed: 11/17/2022] Open
Abstract
Doxorubicin (DOX) is a widely used antineoplastic drug whose efficacy is limited by its cardiotoxicity. The aim of this study was to investigate the possible protective role of the antidiabetic drugs metformin (250 mg/kg dissolved in DW p.o. for seven days) and sitagliptin (10 mg/kg dissolved in DW p.o. for seven days) in a model of DOX-induced (single dose 15 mg/kg i.p. at the fifth day) cardiotoxicity in rats. Results of our study revealed that pretreatment with metformin or sitagliptin produced significant (P < 0.05) cardiac protection manifested by a significant decrease in serum levels of LDH and CK-MB enzymes and cardiac MDA and total nitrites and nitrates levels, a significant increase in cardiac SOD activity, and remarkable improvement in the histopathological features as well as a significant reduction in the immunohistochemical expression of COX-2, iNOS, and caspase-3 enzymes as compared to DOX group. These results may suggest using metformin and/or sitagliptin as preferable drugs for diabetic patients suffering from cancer and receiving DOX in their chemotherapy regimen.
Collapse
|
332
|
Di-Wen S, Pan GZ, Hao L, Zhang J, Xue QZ, Wang P, Yuan QZ. Improved antitumor activity of epirubicin-loaded CXCR4-targeted polymeric nanoparticles in liver cancers. Int J Pharm 2015; 500:54-61. [PMID: 26748365 DOI: 10.1016/j.ijpharm.2015.12.066] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 12/15/2015] [Accepted: 12/27/2015] [Indexed: 10/22/2022]
Abstract
A liver-targeted drug delivery system (CX-EPNP) composed of PLGA/TPGS was prepared and characterized. The surface of nanoparticle was conjugated with LFC131 peptide to increase the specific interaction of carrier with CXCR4 overexpressing liver cancers to enhance the Epirubicin (EPI) delivery to tumors. The particles were nanosized with size than 150 nm and portrayed a sustained release kinetics suggesting its suitability for cancer targeting. The in vitro cell uptake results showed that the introduction of LFC131 to the nanoparticles could increase significantly the affinity to human hepatic carcinoma cells (HepG2) with approximately a 3-fold improvement in cellular uptake than non-targeted one. A specific receptor-mediated uptake was observed by confocal laser scanning microscopy. In addition, CX-EPNP showed remarkable cytotoxicity towards HepG2 cells, and could effectively inhibit tumor growth. The more significant EPI accumulation from CX-EPNP in the cancer cells gave rise to the enhanced EPI cytotoxicity and cell apoptosis. The CX-EPNP distributed mostly in the xenograft tumor after intravenous administration to mice and adequately remained in the blood for at least 24h. It seemed that CX-EPNP upon intravenous injection avoided rapid recognition by Kupffer cells and adequately remained in the blood. These findings suggest that CX-EPNP could effectively inhibit the growth of liver tumors in situ and could potentially reduce the systemic side effects. However, extensive investigation is still needed to assess the possible applications of the CX-EPNP in humans.
Collapse
Affiliation(s)
- Sun Di-Wen
- Department of Hepatobiliary Surgery, Shengli Oilfield Central Hospitial, 31 Jinan Road, Dongying, Shandong Province 257000, PR China
| | - Guo-Zheng Pan
- Department of Hepatobiliary Surgery, Shengli Oilfield Central Hospitial, 31 Jinan Road, Dongying, Shandong Province 257000, PR China
| | - Long Hao
- Department of Hepatobiliary Surgery, Shengli Oilfield Central Hospitial, 31 Jinan Road, Dongying, Shandong Province 257000, PR China
| | - Jian Zhang
- Department of Hepatobiliary Surgery, Shengli Oilfield Central Hospitial, 31 Jinan Road, Dongying, Shandong Province 257000, PR China
| | - Qing-Ze Xue
- Department of Hepatobiliary Surgery, Shengli Oilfield Central Hospitial, 31 Jinan Road, Dongying, Shandong Province 257000, PR China
| | - Peng Wang
- Department of Hepatobiliary Surgery, Shengli Oilfield Central Hospitial, 31 Jinan Road, Dongying, Shandong Province 257000, PR China
| | - Qing-Zhong Yuan
- Department of Hepatobiliary Surgery, Shengli Oilfield Central Hospitial, 31 Jinan Road, Dongying, Shandong Province 257000, PR China.
| |
Collapse
|
333
|
High-Copy Overexpression Screening Reveals PDR5 as the Main Doxorubicin Resistance Gene in Yeast. PLoS One 2015; 10:e0145108. [PMID: 26690737 PMCID: PMC4687100 DOI: 10.1371/journal.pone.0145108] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 11/27/2015] [Indexed: 11/24/2022] Open
Abstract
Doxorubicin is one of the most potent anticancer drugs used in the treatment of various cancer types. The efficacy of doxorubicin is influenced by the drug resistance mechanisms and its cytotoxicity. In this study, we performed a high-copy screening analysis to find genes that play a role in doxorubicin resistance and found several genes (CUE5, AKL1, CAN1, YHR177W and PDR5) that provide resistance. Among these genes, overexpression of PDR5 provided a remarkable resistance, and deletion of it significantly rendered the tolerance level for the drug. Q-PCR analyses suggested that transcriptional regulation of these genes was not dependent on doxorubicin treatment. Additionally, we profiled the global expression pattern of cells in response to doxorubicin treatment and highlighted the genes and pathways that are important in doxorubicin tolerance/toxicity. Our results suggest that many efflux pumps and DNA metabolism genes are upregulated by the drug and required for doxorubicin tolerance.
Collapse
|
334
|
Leukotriene C4 is the major trigger of stress-induced oxidative DNA damage. Nat Commun 2015; 6:10112. [PMID: 26656251 PMCID: PMC4682057 DOI: 10.1038/ncomms10112] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Accepted: 11/04/2015] [Indexed: 12/18/2022] Open
Abstract
Endoplasmic reticulum (ER) stress and major chemotherapeutic agents damage DNA by generating reactive oxygen species (ROS). Here we show that ER stress and chemotherapy induce leukotriene C4 (LTC4) biosynthesis by transcriptionally upregulating and activating the enzyme microsomal glutathione-S-transferase 2 (MGST2) in cells of non-haematopoietic lineage. ER stress and chemotherapy also trigger nuclear translocation of the two LTC4 receptors. Acting in an intracrine manner, LTC4 then elicits nuclear translocation of NADPH oxidase 4 (NOX4), ROS accumulation and oxidative DNA damage. Mgst2 deficiency, RNAi and LTC4 receptor antagonists abolish ER stress- and chemotherapy-induced ROS and oxidative DNA damage in vitro and in mouse kidneys. Cell death and mouse morbidity are also significantly attenuated. Hence, MGST2-generated LTC4 is a major mediator of ER stress- and chemotherapy-triggered oxidative stress and oxidative DNA damage. LTC4 inhibitors, commonly used for asthma, could find broad clinical use in major human pathologies associated with ER stress-activated NOX4. Chemotherapeutic agents elicit ER and oxidative stress as part of their mode of action. Here the authors show that chemotherapy and ER stress trigger MGST2-based biosynthesis of LTC4, whose inhibition abolishes chemotherapy- and ER stress-triggered oxidative stress and DNA damage, resulting in the attenuation of cell death.
Collapse
|
335
|
Vera T, D'Agostino RB, Jordan JH, Whitlock MC, Meléndez GC, Lamar ZS, Porosnicu M, Bonkovsky HL, Poole LB, Hundley WG. Relation of Pre-anthracycline Serum Bilirubin Levels to Left Ventricular Ejection Fraction After Chemotherapy. Am J Cardiol 2015; 116:1752-5. [PMID: 26433273 DOI: 10.1016/j.amjcard.2015.08.042] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 08/21/2015] [Accepted: 08/21/2015] [Indexed: 01/09/2023]
Abstract
Myocardial injury because of oxidative stress manifesting through reductions in left ventricular ejection fraction (LVEF) may occur after the administration of anthracycline-based chemotherapy (A-bC). We hypothesized that bilirubin, an effective endogenous antioxidant, may attenuate the reduction in LVEF that sometimes occurs after receipt of A-bC. We identified 751 consecutively treated patients with cancer who underwent a pre-A-bC LVEF measurement, exhibited a serum total bilirubin level <2 mg/dl, and then received a post-A-bC LVEF assessment because of symptomatology associated with heart failure. Analysis of variance, Tukey's Studentized range test, and chi-square tests were used to evaluate an association between bilirubin and LVEF changes. The LVEF decreased by 10.7 ± 13.7%, 8.9 ± 11.8%, and 7.7 ± 11.5% in group 1 (bilirubin at baseline ≤0.5 mg/dl), group 2 (bilirubin 0.6 to 0.8 mg/dl), and group 3 (bilirubin 0.9 to 1.9 mg/dl), respectively. More group 1 patients experienced >15% decrease in LVEF compared with those in group 3 (p = 0.039). After adjusting for age, coronary artery disease/myocardial infarction, diabetes mellitus, hematocrit, and the use of cardioactive medications, higher precancer treatment bilirubin levels and lesser total anthracycline doses were associated with LVEF preservation (p = 0.047 and 0.011, respectively). In patients treated with anthracyclines who subsequently develop symptoms associated with heart failure, pre-anthracycline treatment serum bilirubin levels inversely correlate with subsequent deterioration in post-cancer treatment LVEF. In conclusion, these results suggest that increased levels of circulating serum total bilirubin, an intrinsic antioxidant, may facilitate preservation of LVEF in patients receiving A-bC for cancer.
Collapse
Affiliation(s)
- Trinity Vera
- Cardiovascular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Ralph B D'Agostino
- Department of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Jennifer H Jordan
- Cardiovascular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Matthew C Whitlock
- Cardiovascular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Giselle C Meléndez
- Cardiovascular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina; Comparative Medicine, Department of Pathology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Zanetta S Lamar
- Hematology and Oncology, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Mercedes Porosnicu
- Hematology and Oncology, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Herbert L Bonkovsky
- Gastroenterology and Hepatology and Molecular Medicine Translational Science, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Leslie B Poole
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, North Carolina; Center for Structural Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - W Gregory Hundley
- Cardiovascular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina; Division of Radiologic Sciences, Department of Radiology, Wake Forest School of Medicine, Winston-Salem, North Carolina.
| |
Collapse
|
336
|
Kwatra M, Kumar V, Jangra A, Mishra M, Ahmed S, Ghosh P, Vohora D, Khanam R. Ameliorative effect of naringin against doxorubicin-induced acute cardiac toxicity in rats. PHARMACEUTICAL BIOLOGY 2015; 54:637-647. [PMID: 26471226 DOI: 10.3109/13880209.2015.1070879] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
CONTEXT Doxorubicin (Dox) is one of the most active chemotherapeutic agents used to treat various types of cancers. Its clinical utility is compromised due to fatal cardiac toxicity characterized by an irreversible cardiomyopathy. OBJECTIVE This study evaluates the cardioprotective potential of naringin (NR) against Dox-induced acute cardiac toxicity in rats. MATERIALS AND METHODS Male Wistar rats were randomly divided into five groups. NR (50 and 100 mg/kg) was administered intraperitoneally (i.p.) daily from 0 to 14 d. Doxorubicin (15 mg/kg, i.p.) was given as a single dose on the 10th day. On the 14th day, all animals were sacrificed and oxidative stress parameters that include malondialdehyde (MDA), glutathione (GSH) level, superoxide dismutase (SOD), catalase (CAT) activities, and all mitochondrial complexes (I-IV) activities were evaluated along with histopathological studies of the heart. RESULTS Doxorubicin-induced cardiotoxicity was confirmed by increased (p < 0.05) MDA, decreased (p < 0.05) GSH levels, SOD, and CAT activities, mitochondrial complexes (I-IV) activities in the heart tissue. NR (100 mg/kg) showed cardioprotection as evident from significant decreased MDA (p < 0.001) level, raised (p < 0.001) GSH level, SOD and CAT activities and increased mitochondrial complexes I (p < 0.01), II (p < 0.001), III (p < 0.001), and IV (p < 0.05) activities. Further, Dox-induced cardiotoxicity was confirmed by histopathological studies. These obtained results indicated the protective role of NR against Dox-induced cardiac toxicity in rats. CONCLUSION NR can be used in combination with Dox due to its high cardioprotective effect against Dox-induced cardiomyopathy.
Collapse
Affiliation(s)
- Mohit Kwatra
- a Pharmacology Research Laboratory, Department of Pharmacology, Faculty of Pharmacy, Jamia Hamdard (Hamdard University) , New Delhi , India
| | - Vikas Kumar
- a Pharmacology Research Laboratory, Department of Pharmacology, Faculty of Pharmacy, Jamia Hamdard (Hamdard University) , New Delhi , India
| | - Ashok Jangra
- b Department of Pharmacology and Toxicology , National Institute of Pharmaceutical Education and Research Guwahati , Guwahati , Assam , India
| | - Murli Mishra
- c Department of Toxicology and Cancer Biology , College of Medicine, University of Kentucky , Lexington , KY , USA
| | - Sahabuddin Ahmed
- b Department of Pharmacology and Toxicology , National Institute of Pharmaceutical Education and Research Guwahati , Guwahati , Assam , India
| | - Pinaki Ghosh
- d Department of Pharmacology , Bharati Vidyapeeth University, Poona College of Pharmacy , Erandwane , Pune , Maharashtra , India , and
| | - Divya Vohora
- a Pharmacology Research Laboratory, Department of Pharmacology, Faculty of Pharmacy, Jamia Hamdard (Hamdard University) , New Delhi , India
| | - Razia Khanam
- a Pharmacology Research Laboratory, Department of Pharmacology, Faculty of Pharmacy, Jamia Hamdard (Hamdard University) , New Delhi , India
- e Department of Pharmacology , Gulf Medical University , Ajman , United Arab Emirates
| |
Collapse
|
337
|
Angsutararux P, Luanpitpong S, Issaragrisil S. Chemotherapy-Induced Cardiotoxicity: Overview of the Roles of Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:795602. [PMID: 26491536 PMCID: PMC4602327 DOI: 10.1155/2015/795602] [Citation(s) in RCA: 178] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 05/17/2015] [Indexed: 02/02/2023]
Abstract
Chemotherapy-induced cardiotoxicity is a serious complication that poses a serious threat to life and limits the clinical use of various chemotherapeutic agents, particularly the anthracyclines. Understanding molecular mechanisms of chemotherapy-induced cardiotoxicity is a key to effective preventive strategies and improved chemotherapy regimen. Although no reliable and effective preventive treatment has become available, numerous evidence demonstrates that chemotherapy-induced cardiotoxicity involves the generation of reactive oxygen species (ROS). This review provides an overview of the roles of oxidative stress in chemotherapy-induced cardiotoxicity using doxorubicin, which is one of the most effective chemotherapeutic agents against a wide range of cancers, as an example. Current understanding in the molecular mechanisms of ROS-mediated cardiotoxicity will be explored and discussed, with emphasis on cardiomyocyte apoptosis leading to cardiomyopathy. The review will conclude with perspectives on model development needed to facilitate further progress and understanding on chemotherapy-induced cardiotoxicity.
Collapse
Affiliation(s)
- Paweorn Angsutararux
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Sudjit Luanpitpong
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Surapol Issaragrisil
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| |
Collapse
|
338
|
Abu-Elsaad NM, Abd Elhameed AG, El-Karef A, Ibrahim TM. Yogurt Containing the ProbacteriaLactobacillus acidophilusCombined with Natural Antioxidants Mitigates Doxorubicin-Induced Cardiomyopathy in Rats. J Med Food 2015; 18:950-9. [DOI: 10.1089/jmf.2014.0104] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Affiliation(s)
- Nashwa M. Abu-Elsaad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Ahmed G. Abd Elhameed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Amr El-Karef
- Department of Pathology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Tarek M. Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|
339
|
Jin L, Zhao X, Qin Y, Zhu W, Zhang W, Liu A, Luo Z. Soy isoflavones protect against H₂O₂-induced injury in human umbilical vein endothelial cells. Mol Med Rep 2015; 12:4476-4482. [PMID: 26095641 DOI: 10.3892/mmr.2015.3949] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Accepted: 05/19/2015] [Indexed: 11/05/2022] Open
Abstract
The aim of this study was to investigate the effects of soy isoflavones on the injury of human umbilical vein endothelial cells induced by H2O2. EVC‑304 cells were preincubated with soy isoflavones for 12 h, and then exposed to 100 µM H2O2 for 1 h. Cell viability was evaluated by a 3‑(4,5‑di‑methylthiazol‑2‑yl) 2,5‑diphenyltetrazolium bromide assay. The apoptosis of EVC‑304 cells was detected by Hoechst 33258 and Annexin‑V/propidium iodide staining. The oxidative stress‑related biochemical parameters were detected and the expression of apoptosis‑related proteins was examined by western blot analysis. The results showed that incubation with soy isoflavones caused a significant increase in the viability of EVC‑304 cells and a decrease in cell apoptosis induced by H2O2. Soy isoflavones also markedly enhanced the activities of superoxide dismutase and glutathione peroxidase, and reduced the level of malondialdehyde. Western blot analysis results show that soy isoflavones can modulate the activation of nuclear factor‑κB and the mitochondria‑mediated apoptosis signaling pathway. The results of this study indicated the potential biological relevance of soy isoflavones in the therapy of cardiovascular diseases.
Collapse
Affiliation(s)
- Lianhai Jin
- Department of Psychology, Jilin Medical College, Jilin 132013, P.R. China
| | - Xingyu Zhao
- Department of Psychology, Jilin Medical College, Jilin 132013, P.R. China
| | - Yingxin Qin
- Department of Psychology, Jilin Medical College, Jilin 132013, P.R. China
| | - Wenhe Zhu
- Department of Psychology, Jilin Medical College, Jilin 132013, P.R. China
| | - Wei Zhang
- Department of Psychology, Jilin Medical College, Jilin 132013, P.R. China
| | - Anheng Liu
- Department of Cardiology, 307 Hospital of PLA, Academy of Military Medical Sciences, Beijing 100071, P.R. China
| | - Zhengli Luo
- Department of Psychology, Jilin Medical College, Jilin 132013, P.R. China
| |
Collapse
|
340
|
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: 3.1] [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
|
341
|
Ovatodiolide sensitizes aggressive breast cancer cells to doxorubicin, eliminates their cancer stem cell-like phenotype, and reduces doxorubicin-associated toxicity. Cancer Lett 2015; 364:125-34. [DOI: 10.1016/j.canlet.2015.05.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 02/26/2015] [Accepted: 05/04/2015] [Indexed: 01/18/2023]
|
342
|
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
|
343
|
Increased Oxidative Stress as a Selective Anticancer Therapy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:294303. [PMID: 26273420 PMCID: PMC4529973 DOI: 10.1155/2015/294303] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 02/11/2015] [Indexed: 12/18/2022]
Abstract
Reactive oxygen species (ROS) are closely related to tumorgenesis. Under hypoxic environment, increased levels of ROS induce the expression of hypoxia inducible factors (HIFs) in cancer stem cells (CSCs), resulting in the promotion of the upregulation of CSC markers, and the reduction of intracellular ROS level, thus facilitating CSCs survival and proliferation. Although the ROS level is regulated by powerful antioxidant defense mechanisms in cancer cells, it is observed to remain higher than that in normal cells. Cancer cells may be more sensitive than normal cells to the accumulation of ROS; consequently, it is supposed that increased oxidative stress by exogenous ROS generation therapy has an effect on selectively killing cancer cells without affecting normal cells. This paper reviews the mechanisms of redox regulation in CSCs and the pivotal role of ROS in anticancer treatment.
Collapse
|
344
|
Emer E, Yildiz O, Seyrek M, Demirkol S, Topal T, Kurt B, Sayal A. High-dose testosterone and dehydroepiandrosterone induce cardiotoxicity in rats. Hum Exp Toxicol 2015. [DOI: 10.1177/0960327115595706] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The aim of this study is to assess cardiotoxic effect of testosterone (TES) and dehydroepiandrosterone (DHEA) in Sprague Dawley rats. We compared the impact of subacute (14 days) and subchronic (90 days) administration of suprapharmacologic doses of TES and DHEA on body weight, locomotor activity, muscle strength, echocardiographic parameters, heart histopathology, and oxidative stress markers with the control group. Testosterone (10, 30, and 100 mg/100 g body weight) and DHEA (10 mg/100 g body weight) administration decreased the body weights and locomotor activity ( p < 0.05), and the combination of both increased muscle strength ( p < 0.05) in rats. In our histopathological evaluation, misshapen cell nuclei, disorganized myocardial fibers, and leukocytic infiltrates were observed in high-dose TES (100 mg/100 g)-treated rats, especially on day 14. On day 90, mild changes such as misshapen cell nuclei, disorganized myocardial fibers, and leukocytic infiltrates were observed in TES and DHEA-treated groups. According to our echocardiographic study on day 14 and day 90, TES, especially at high doses, induced increase in left ventricular posterior wall diameter and ejection fraction ( p < 0.05). In this study, blood oxidative stress marker malondialdehyde was increased slightly but not significantly in TES and DHEA groups. On the other hand, antioxidant enzymes such as SOD and glutathione peroxidase (GSH-Px) levels were slightly but not significantly increased in TES and DHEA groups. These data demonstrate that the potential risk to cardiac health due to exogenous androgen use may be related to oxidative stress in rats.
Collapse
Affiliation(s)
- E Emer
- Institute of Forensic Sciences, Ankara University, Dikimevi-Ankara, Turkey
| | - O Yildiz
- Department of Pharmacology, Gülhane Faculty of Medicine, Ankara, Turkey
| | - M Seyrek
- Department of Pharmacology, Gülhane Faculty of Medicine, Ankara, Turkey
| | - S Demirkol
- Department of Cardiology, Gülhane Faculty of Medicine, Ankara, Turkey
| | - T Topal
- Department of Physiology, Gülhane Faculty of Medicine, Ankara, Turkey
| | - B Kurt
- Department of Pathology, Gülhane Faculty of Medicine, Ankara, Turkey
| | - A Sayal
- Center of Pharmaceutical Sciences, Gülhane Medical Academy, Ankara, Turkey
| |
Collapse
|
345
|
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: 1.0] [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
|
346
|
Abdel-Rahman O, Alorabi M. Use of angiotensin-converting enzyme inhibitors in the prophylaxis of anthracycline or trastuzumab-related cardiac dysfunction: preclinical and clinical considerations. Expert Rev Anticancer Ther 2015; 15:829-837. [DOI: 10.1586/14737140.2015.1047766] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|
347
|
Wei L, Surma M, Gough G, Shi S, Lambert-Cheatham N, Chang J, Shi J. Dissecting the Mechanisms of Doxorubicin and Oxidative Stress-Induced Cytotoxicity: The Involvement of Actin Cytoskeleton and ROCK1. PLoS One 2015; 10:e0131763. [PMID: 26134406 PMCID: PMC4489912 DOI: 10.1371/journal.pone.0131763] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 06/05/2015] [Indexed: 01/13/2023] Open
Abstract
We have recently reported that ROCK1 deficiency in mouse embryonic fibroblasts (MEF) has superior anti-apoptotic and pro-survival effects than antioxidants against doxorubicin, a chemotherapeutic drug. Although oxidative stress is the most widely accepted mechanism, our studies suggest that ROCK1-dependent actin cytoskeleton remodeling plays a more important role in mediating doxorubicin cytotoxicity on MEFs. To further explore the contributions of ROCK1-dependent actin cytoskeleton remodeling in response to stress, this study investigates the mechanistic differences between the cytotoxic effects of doxorubicin versus hydrogen peroxide (H2O2), with a focus on cytoskeleton alterations, apoptosis and necrosis induction. We found that both types of stress induce caspase activation but with different temporal patterns and magnitudes in MEFs: H2O2 induces the maximal levels (2 to 4-fold) of activation of caspases 3, 8, and 9 within 4 h, while doxorubicin induces much higher maximal levels (15 to 25-fold) of caspases activation at later time points (16-24 h). In addition, necrosis induced by H2O2 reaches maximal levels within 4 h while doxorubicin-induced necrosis largely occurs at 16-24 h secondary to apoptosis. Moreover, both types of stress induce actin cytoskeleton remodeling but with different characteristics: H2O2 induces disruption of stress fibers associated with cytosolic translocation of phosphorylated myosin light chain (p-MLC) from stress fibers, while doxorubicin induces cortical F-actin formation associated with cortical translocation of p-MLC from central stress fibers. Furthermore, N-acetylcysteine (an antioxidant) is a potent suppressor for H2O2-induced cytotoxic effects including caspase activation, necrosis, and cell detachment, but shows a much reduced inhibition on doxorubicin-induced changes. On the other hand, ROCK1 deficiency is a more potent suppressor for the cytotoxic effects induced by doxorubicin than by H2O2. These results support the notion that doxorubicin induces caspase activation, necrosis, and actin cytoskeleton alterations largely through ROCK1-dependent and oxidative stress-independent pathways.
Collapse
Affiliation(s)
- Lei Wei
- Riley Heart Research Center, Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University, School of Medicine, Indianapolis, Indiana, United States of America
- * E-mail: (LW); (JS)
| | - Michelle Surma
- Riley Heart Research Center, Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University, School of Medicine, Indianapolis, Indiana, United States of America
| | - Gina Gough
- Riley Heart Research Center, Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University, School of Medicine, Indianapolis, Indiana, United States of America
| | - Stephanie Shi
- Riley Heart Research Center, Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University, School of Medicine, Indianapolis, Indiana, United States of America
| | - Nathan Lambert-Cheatham
- Riley Heart Research Center, Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University, School of Medicine, Indianapolis, Indiana, United States of America
| | - Jiang Chang
- Texas A&M University Health Science Center, Institute of Biosciences and Technology, Houston, Texas, United States of America
| | - Jianjian Shi
- Riley Heart Research Center, Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University, School of Medicine, Indianapolis, Indiana, United States of America
- * E-mail: (LW); (JS)
| |
Collapse
|
348
|
Rahmatollahi M, Baram SM, Rahimian R, Saeedi Saravi SS, Dehpour AR. Peroxisome Proliferator-Activated Receptor-α Inhibition Protects Against Doxorubicin-Induced Cardiotoxicity in Mice. Cardiovasc Toxicol 2015; 16:244-50. [DOI: 10.1007/s12012-015-9332-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
349
|
Affiliation(s)
- Preeti Kumari
- Department of Pharmacy, Birla Institute of Technology and Science – Pilani, Hyderabad Campus, Hyderabad, Andhra Pradesh, India
| | - Balaram Ghosh
- Department of Pharmacy, Birla Institute of Technology and Science – Pilani, Hyderabad Campus, Hyderabad, Andhra Pradesh, India
| | - Swati Biswas
- Department of Pharmacy, Birla Institute of Technology and Science – Pilani, Hyderabad Campus, Hyderabad, Andhra Pradesh, India
| |
Collapse
|
350
|
High Throughput Screening Identifies a Novel Compound Protecting Cardiomyocytes from Doxorubicin-Induced Damage. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:178513. [PMID: 26137186 PMCID: PMC4475553 DOI: 10.1155/2015/178513] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 04/27/2015] [Accepted: 05/20/2015] [Indexed: 12/01/2022]
Abstract
Antracyclines are effective antitumor agents. One of the most commonly used antracyclines is doxorubicin, which can be successfully used to treat a diverse spectrum of tumors. Application of these drugs is limited by their cardiotoxic effect, which is determined by a lifetime cumulative dose. We set out to identify by high throughput screening cardioprotective compounds protecting cardiomyocytes from doxorubicin-induced injury. Ten thousand compounds of ChemBridge's DIVERSet compound library were screened to identify compounds that can protect H9C2 rat cardiomyocytes against doxorubicin-induced cell death. The most effective compound proved protective in doxorubicin-treated primary rat cardiomyocytes and was further characterized to demonstrate that it significantly decreased doxorubicin-induced apoptotic and necrotic cell death and inhibited doxorubicin-induced activation of JNK MAP kinase without having considerable radical scavenging effect or interfering with the antitumor effect of doxorubicin. In fact the compound identified as 3-[2-(4-ethylphenyl)-2-oxoethyl]-1,2-dimethyl-1H-3,1-benzimidazol-3-ium bromide was toxic to all tumor cell lines tested even without doxorubicine treatment. This benzimidazole compound may lead, through further optimalization, to the development of a drug candidate protecting the heart from doxorubicin-induced injury.
Collapse
|