151
|
Uygur R, Aktas C, Tulubas F, Alpsoy S, Topcu B, Ozen OA. Cardioprotective effects of fish omega-3 fatty acids on doxorubicin-induced cardiotoxicity in rats. Hum Exp Toxicol 2013; 33:435-45. [DOI: 10.1177/0960327113493304] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The aim of this study was to investigate the protective effects of fish omega-3 (n-3) fatty acids on doxorubicin (DOX)-induced acute cardiotoxicity. A total of 24 rats were divided into three groups: control, DOX-treated, and DOX treated with fish n-3 fatty acids. Control group received 0.4 ml/kg/day of saline intragastrically. The rats in the fish n-3 fatty acid-pretreated group were given 400 mg/kg/day fish n-3 fatty acids for 30 days by intragastric intubation. To induce acute cardiotoxicity, DOX (30 mg/kg) was injected intraperitoneally by a single dose and the rats were killed after 48 h. DOX treatment caused severe damage in heart tissues. Disorganization of myocardial muscle fibers, myofibrillar loss, and cardiotoxic myocardial fibers with cytoplasmic vacuoles were seen. Fish n-3 fatty acid-treated rats showed an improved histological appearance in the DOX-treated group. Our data indicate a significant reduction in the activity of terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end labeling in cardiomyocytes of the DOX-treated group with fish n-3 fatty acids therapy. The DOX-treated with fish n-3 fatty acids group showed a significant decrease in malondialdehyde levels, and an increase in superoxide dismutase and glutathione peroxidase activities in comparison with the DOX-treated group. This study showed that fish n-3 fatty acids may be a suitable cardioprotector against acute toxic effects of DOX.
Collapse
Affiliation(s)
- R Uygur
- Department of Anatomy, Faculty of Medicine, Namik Kemal University, Tekirdag, Turkey
| | - C Aktas
- Department of Histology and Embryology, Faculty of Medicine, Namik Kemal University, Tekirdag, Turkey
| | - F Tulubas
- Department of Biochemistry, Faculty of Medicine, Namik Kemal University, Tekirdag, Turkey
| | - S Alpsoy
- Department of Cardiology, Faculty of Medicine, Namik Kemal University, Tekirdag, Turkey
| | - B Topcu
- Department of Biostatistics, Faculty of Medicine, Namik Kemal University, Tekirdag, Turkey
| | - OA Ozen
- Department of Anatomy, Faculty of Medicine, Namik Kemal University, Tekirdag, Turkey
| |
Collapse
|
152
|
Lee CC, Chang DM, Huang KF, Chen CL, Chen TC, Lo Y, Guh JH, Huang HS. Design, synthesis and antiproliferative evaluation of fluorenone analogs with DNA topoisomerase I inhibitory properties. Bioorg Med Chem 2013; 21:7125-33. [PMID: 24094433 DOI: 10.1016/j.bmc.2013.09.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Revised: 09/03/2013] [Accepted: 09/03/2013] [Indexed: 11/29/2022]
Abstract
A series of 2,7-diamidofluorenones were designed, synthesized, and screened by SRB assay. Some synthesized compounds exhibited antitumor activities in submicromolar range. Ten compounds (3a, 3b, 3c, 3g, 3j, 3l, 4a, 4h, 4i, and 4j) were also selected by NCI screening system and 3c (GI50=1.66 μM) appeared to be the most active agent of this series. Furthermore, 3c attenuated topoisomerase I-mediated DNA relaxation at low micromolar concentrations. These results indicated that fluorenones have potential to be further developed into anticancer drugs.
Collapse
Affiliation(s)
- Chia-Chung Lee
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan; School of Pharmacy, National Defense Medical Center, Taipei 114, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
153
|
Mazevet M, Moulin M, Llach-Martinez A, Chargari C, Deutsch E, Gomez AM, Morel E. Complications of chemotherapy, a basic science update. Presse Med 2013; 42:e352-61. [PMID: 23972551 DOI: 10.1016/j.lpm.2013.06.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Accepted: 06/12/2013] [Indexed: 11/25/2022] Open
Abstract
Anthracyclines, discovered 50 years ago, are antibiotics widely used as antineoplastic agents and are among the most successful anticancer therapies ever developed to treat a wide range of cancers, including hematological malignancies, soft tissue sarcomas and solid tumors. However, some anthracyclines, including doxorubicin, exhibit major signs of cardiotoxicity that may ultimately lead to heart failure (HF). Despite intensive research on doxorubicine-induced cardiotoxicity, the underlying mechanisms responsible for doxorubicin-induced cardiotoxicity have not been fully elucidated yet. Published literature so far has focused mostly on mitochondria dysfunction with consequent oxidative stress, Ca(2+) overload, and cardiomyocyte death as doxorubicin side effects, leading to heart dysfunction. This review focuses on the current understanding of the molecular mechanisms underlying doxorubicin-induced cardiomyocyte death (i.e.: cardiomyocyte death, mitochondria metabolism and bioenergetic alteration), but we will also point to new directions of possible mechanisms, suggesting potent prior or concomitant alterations of specific signaling pathways with molecular actors directly targeted by the anticancer drugs itself (i.e. calcium homeostasis or cAMP signaling cascade). The mechanisms of anticancer cardiac toxicity may be more complex than just mitochondria dysfunction. Partnership of both basic and clinical research is needed to promote new strategies in diagnosis, therapies with concomitant cardioprotection in order to achieve cancer treatment with acceptable cardiotoxicity along life span.
Collapse
Affiliation(s)
- Marianne Mazevet
- Inserm UMR-S 769, LabEx Lermit-DHU Torino, université Paris-Sud, faculté de pharmacie, signalisation et physiopathologie cardiaque, 92296 Châtenay-Malabry cedex, France
| | | | | | | | | | | | | |
Collapse
|
154
|
Hanušová V, Tomšík P, Kriesfalusyová L, Pakostová A, Boušová I, Skálová L. In vivo effect of oracin on doxorubicin reduction, biodistribution and efficacy in Ehrlich tumor bearing mice. Pharmacol Rep 2013; 65:445-52. [PMID: 23744429 DOI: 10.1016/s1734-1140(13)71020-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2011] [Revised: 11/26/2012] [Indexed: 01/22/2023]
Abstract
BACKGROUND The limitation of carbonyl reduction represents one possible way to increase the effectiveness of anthracycline doxorubicin (DOX) in cancer cells and decrease its toxicity in normal cells. In vitro, isoquinoline derivative oracin (ORC) inhibited DOX reduction and increased the antiproliferative effect of DOX in MCF-7 breast cancer cells. Moreover, ORC significantly decreases DOX toxicity in non-cancerous MCF-10A breast cells and in hepatocytes. The present study was designed to test in mice the in vivo effect of ORC on plasma and tissue concentrations of DOX and its main metabolite DOXOL. The effect of ORC on DOX efficacy in mice bearing solid Ehrlich tumors (EST) was also studied. METHODS DOX and DOX + ORC combinations were iv administered to healthy mice. Blood samples, livers and hearts were collected during the following 48 h. DOX and DOXOL concentrations were assayed using HPLC. The mice with inoculated EST cells were treated repeatedly iv with DOX and DOX + ORC combinations, and the growth of tumors was monitored. RESULTS ORC in combination with DOX significantly decreased DOXOL plasma concentrations during four hours after administration, but this significantly affected neither DOX plasma concentrations nor DOX or DOXOL concentrations in the liver and heart at any of intervals tested. In EST bearing mice, ORC did not significantly affect DOX efficacy on tumor growth. However, EST was shown to be an improper model for the testing of ORC efficacy in vivo, as ORC did not inhibit DOXOL formation in EST. CONCLUSIONS In vivo, ORC was able to retard DOXOL formation but was not able to improve DOX efficacy in EST-bearing mice.
Collapse
Affiliation(s)
- Veronika Hanušová
- Department of Biochemical Sciences, Faculty of Pharmacy, Charles University in Prague, Heyrovského 1203, Hradec Králové, CZ 500 05, Czech Republic
| | | | | | | | | | | |
Collapse
|
155
|
Jin Z, Zhang J, Zhi H, Hong B, Zhang S, Guo H, Li L. Beneficial effects of tadalafil on left ventricular dysfunction in doxorubicin-induced cardiomyopathy. J Cardiol 2013; 62:110-6. [PMID: 23731918 DOI: 10.1016/j.jjcc.2013.03.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Revised: 03/14/2013] [Accepted: 03/21/2013] [Indexed: 11/15/2022]
Abstract
BACKGROUND It is not clear yet how tadalafil affects nonischemic cardiomyopathy, although its beneficial effects on acute myocardial infarction are well-known. We investigated tadalafil's beneficial effects on nonischemic cardiomyopathy and the specific mechanisms of its effects. METHODS Cardiomyopathy was induced in mice by a single intraperitoneal injection of doxorubicin (15 mg/kg). In some cases, tadalafil (4 mg/kg/day, p.o., 14 days) was started simultaneously. After two weeks, cardiac function was evaluated by echocardiography and cardiac catheterization, then all of the mice were killed and cardiac specimens were subjected for hemotoxylin and eosin staining, Masson's trichrome staining, terminal deoxynucleotidyltransferase dUTP nick-end labeling assay, enzyme-linked immunosorbent assay, and Western blot. RESULTS Two weeks later, left ventricular dilatation and dysfunction were apparent in mice given doxorubicin but were significantly attenuated by tadalafil treatment. Tadalafil also protected hearts against doxorubicin-induced cardiomyocyte atrophy/degeneration and myocardial fibrosis. No doxorubicin-induced apoptotic effects were seen between groups. Cardiac cGMP level was lower in the doxorubicin-treated group, however it was significantly increased with tadalafil treatment. Compared to the control group, the myocardial expression of 3 sarcomeric proteins, myosin heavy chain, troponin I, and desmin were significantly decreased in the doxorubicin-treated group, which were restored by the tadalafil treatment. CONCLUSIONS The present study indicates a protective effect of tadalafil mainly through cGMP signaling pathway against doxorubicin-induced nonischemic cardiomyopathy.
Collapse
Affiliation(s)
- Zhe Jin
- Department of Cardiology, The Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | | | | | | | | | | | | |
Collapse
|
156
|
Nagendran J, Kienesberger PC, Pulinilkunnil T, Zordoky BN, Sung MM, Kim T, Young ME, Dyck JRB. Cardiomyocyte specific adipose triglyceride lipase overexpression prevents doxorubicin induced cardiac dysfunction in female mice. Heart 2013; 99:1041-7. [PMID: 23704323 DOI: 10.1136/heartjnl-2013-303843] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES Anthracyclines such as doxorubicin are an effective class of antineoplastic agents. Despite its efficacy in the treatment of a variety of cancers, the clinical use of doxorubicin is limited by cardiac side effects. While it has been suggested that doxorubicin alters myocardial fatty acid metabolism, it is poorly understood whether this is the case and whether variations in myocardial triacylglycerol (TAG) metabolism contribute to doxorubicin induced cardiotoxicity. Since TAG catabolism in the heart is controlled by adipose triglyceride lipase (ATGL), this study examined the influence of doxorubicin on cardiac energy metabolism and TAG values as well as the consequence of forced expression of ATGL in the setting of doxorubicin induced cardiotoxicity. DESIGN AND SETTING Wild type (WT) mice and mice with cardiomyocyte specific ATGL overexpression were divided into two groups per genotype that received a weekly intraperitoneal injection of saline or doxorubicin for 4 weeks. RESULTS Four weeks of doxorubicin administration significantly impaired in vivo systolic function (11% reduction in ejection fraction, p<0.05), which was associated with increased lung wet to dry weight ratios. Furthermore, doxorubicin induced cardiac dysfunction was independent of changes in glucose and fatty acid oxidation in WT hearts. However, doxorubicin administration significantly reduced myocardial TAG content in WT mice (p<0.05). Importantly, cardiomyocyte specific ATGL overexpression and the resulting decrease in cardiac TAG accumulation attenuated the decrease in ejection fraction (p<0.05) and thus protected mice from doxorubicin induced cardiac dysfunction. CONCLUSIONS Taken together, our data suggest that chronic reduction in myocardial TAG content by cardiomyocyte specific ATGL overexpression is able to prevent doxorubicin induced cardiac dysfunction.
Collapse
|
157
|
Smuder AJ, Kavazis AN, Min K, Powers SK. Doxorubicin-induced markers of myocardial autophagic signaling in sedentary and exercise trained animals. J Appl Physiol (1985) 2013; 115:176-85. [PMID: 23703114 DOI: 10.1152/japplphysiol.00924.2012] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Doxorubicin (DOX) is an effective antitumor agent used in cancer treatment. However, its clinical use is limited due to cardiotoxicity. Indeed, the side effects of DOX are irreversible and include the development of cardiomyopathy and ultimately congestive heart failure. Although many studies have investigated the events leading to DOX-induced cardiotoxicity, the mechanisms responsible for DOX-induced cardiotoxicity remain unknown. In general, evidence suggests that DOX-induced cardiotoxicity is associated with an increased generation of reactive oxygen species and oxidative damage, leading to the activation of cellular proteolytic systems. In this regard, the autophagy/lysosomal proteolytic system is a constitutively active catabolic process that is responsible for the degradation of both organelles and cytosolic proteins. We tested the hypothesis that systemic DOX administration results in altered cardiac gene and protein expression of mediators of the autophagy/lysosomal system. Our results support this hypothesis, as DOX treatment increased both the mRNA and protein levels of numerous key autophagy genes. Because exercise training has been shown to be cardioprotective against DOX-induced damage, we also determined whether exercise training before DOX administration alters the expression of important components of the autophagy/lysosomal system in cardiac muscle. Our findings show that exercise training inhibits DOX-induced cardiac increases in autophagy signaling. Collectively, our results reveal that DOX administration promotes activation of the autophagy/lysosomal system pathway in the heart, and that endurance exercise training can be a cardioprotective intervention against myocardial DOX-induced toxicity.
Collapse
Affiliation(s)
- Ashley J Smuder
- Department of Applied Physiology and Kinesiology, Center for Exercise Science, University of Florida, Gainesville, Florida 32611, USA.
| | | | | | | |
Collapse
|
158
|
Volkova M, Russell R. Anthracycline cardiotoxicity: prevalence, pathogenesis and treatment. Curr Cardiol Rev 2013; 7:214-20. [PMID: 22758622 PMCID: PMC3322439 DOI: 10.2174/157340311799960645] [Citation(s) in RCA: 584] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Revised: 09/15/2011] [Accepted: 12/02/2011] [Indexed: 11/22/2022] Open
Abstract
Anthracyclines, such as doxorubicin and idarubicin, remain an important class of chemotherapeutic agents. Unfortunately, their efficacy in treating cancer is limited by a cumulative dose-dependent cardiotoxicity, which can cause irreversible heart failure. In this review, we discuss the pathogenesis and incidence of anthracycline-induced cardiotoxicity as well as methods to detect, prevent and treat the condition.
Collapse
Affiliation(s)
- Maria Volkova
- Section of Cardiovascular Medicine, Yale University School of Medicine, USA
| | | |
Collapse
|
159
|
Bacterial inactivation of the anticancer drug doxorubicin. ACTA ACUST UNITED AC 2013; 19:1255-64. [PMID: 23102220 DOI: 10.1016/j.chembiol.2012.08.011] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Revised: 07/10/2012] [Accepted: 08/06/2012] [Indexed: 11/23/2022]
Abstract
Microbes are exposed to compounds produced by members of their ecological niche, including molecules with antibiotic or antineoplastic activities. As a result, even bacteria that do not produce such compounds can harbor the genetic machinery to inactivate or degrade these molecules. Here, we investigated environmental actinomycetes for their ability to inactivate doxorubicin, an aminoglycosylated anthracycline anticancer drug. One strain, Streptomyces WAC04685, inactivates doxorubicin via a deglycosylation mechanism. Activity-based purification of the enzymes responsible for drug inactivation identified the NADH dehydrogenase component of respiratory electron transport complex I, which was confirmed by gene inactivation studies. A mechanism where reduction of the quinone ring of the anthracycline by NADH dehydrogenase leads to deglycosylation is proposed. This work adds anticancer drug inactivation to the enzymatic inactivation portfolio of actinomycetes and offers possibilities for novel applications in drug detoxification.
Collapse
|
160
|
Carvalho FS, Burgeiro A, Garcia R, Moreno AJ, Carvalho RA, Oliveira PJ. Doxorubicin-Induced Cardiotoxicity: From Bioenergetic Failure and Cell Death to Cardiomyopathy. Med Res Rev 2013; 34:106-35. [DOI: 10.1002/med.21280] [Citation(s) in RCA: 349] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Filipa S. Carvalho
- CNC-Center for Neuroscience and Cell Biology; University of Coimbra; 3004-517 Coimbra Portugal
- Department of Life Sciences; University of Coimbra; 3004-517 Coimbra Portugal
| | - Ana Burgeiro
- CNC-Center for Neuroscience and Cell Biology; University of Coimbra; 3004-517 Coimbra Portugal
- IMAR-Institute of Marine Research; University of Coimbra; Portugal
| | - Rita Garcia
- IMAR-Institute of Marine Research; University of Coimbra; Portugal
| | - António J. Moreno
- Department of Life Sciences; University of Coimbra; 3004-517 Coimbra Portugal
- IMAR-Institute of Marine Research; University of Coimbra; Portugal
| | - Rui A. Carvalho
- CNC-Center for Neuroscience and Cell Biology; University of Coimbra; 3004-517 Coimbra Portugal
- Department of Life Sciences; University of Coimbra; 3004-517 Coimbra Portugal
| | - Paulo J. Oliveira
- CNC-Center for Neuroscience and Cell Biology; University of Coimbra; 3004-517 Coimbra Portugal
| |
Collapse
|
161
|
Morrissy S, Strom J, Purdom-Dickinson S, Chen QM. NAD(P)H:quinone oxidoreductase 1 is induced by progesterone in cardiomyocytes. Cardiovasc Toxicol 2013; 12:108-14. [PMID: 21947872 DOI: 10.1007/s12012-011-9144-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
NAD(P)H: quinone oxidoreductase 1 (NQO1) is a ubiquitous flavoenzyme that catalyzes two-electron reduction of various quinones by utilizing NAD(P)H as an electron donor. Our previous study found that progesterone (PG) can protect cardiomyocytes from apoptosis induced by doxorubicin (Dox). Microarray analyses of genes induced by PG had led to the discovery of induction of NQO1 mRNA. We report here that PG induces NQO1 protein and its activity in a dose-dependent manner. Whereas NQO1 is well known as a target gene of Nrf2 transcription factor due to the presence of antioxidant response element (ARE) in the promoter, PG did not activate the ARE, suggesting Nrf2-independent induction of NQO1. To address the role of NQO1 induction in PG-induced cytoprotection, we tested the effect of NQO1 inducer β-naphthoflavone and inhibitor dicoumarol. Induction of NQO1 by β-naphthoflavone decreased Dox-induced apoptosis and potentiated the protective effect of PG as measured by caspase-3 activity. PG-induced NQO1 activity was inhibited with dicoumarol, which did not affect PG-induced cytoprotection. Dicoumarol treatment alone potentiated Dox-induced caspase-3 activity. These data suggest that while NQO1 plays a role in PG-induced cytoprotection, there are additional components contributing to PG-induced cytoprotection.
Collapse
Affiliation(s)
- Stephen Morrissy
- Department of Pharmacology, College of Medicine, University of Arizona, 1501 N. Campbell Ave, Tucson, AZ 85724, USA
| | | | | | | |
Collapse
|
162
|
Tang Y, McGoron AJ. Increasing the rate of heating: A potential therapeutic approach for achieving synergistic tumour killing in combined hyperthermia and chemotherapy. Int J Hyperthermia 2013; 29:145-55. [DOI: 10.3109/02656736.2012.760757] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
163
|
Guo R, Lin J, Xu W, Shen N, Mo L, Zhang C, Feng J. Hydrogen sulfide attenuates doxorubicin-induced cardiotoxicity by inhibition of the p38 MAPK pathway in H9c2 cells. Int J Mol Med 2013; 31:644-50. [PMID: 23338126 DOI: 10.3892/ijmm.2013.1246] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2012] [Accepted: 12/04/2012] [Indexed: 11/06/2022] Open
Abstract
We previously demonstrated the protective effect of hydrogen sulfide (H2S) against doxorubicin (DOX)-induced cardiotoxicity through inhibition of endoplasmic reticulum stress. The aim of the present study was to explore the role of p38 mitogen-activated protein kinase (MAPK) in DOX-induced cardiotoxicity and ascertain whether exogenous H2S protects DOX-induced injury by inhibiting p38 MAPK in cardiomyoblasts (H9c2). We observed that exposure of H9c2 cells to 5 µM DOX not only markedly induced injuries, including cytotoxicity, apoptosis, overproduction of reactive oxygen species (ROS) and dissipation of mitochondrial membrane potential (MMP), but also enhanced the expression level of phosphorylated (p)-p38 MAPK. The DOX-induced increase in expression of p-p38 MAPK was significantly attenuated by pretreatment of H9c2 cells with either 400 µM sodium hydrogen sulfide (NaHS) (a donor of H2S) or 1,000 µM N-acetyl-L-cysteine (NAC, an ROS scavenger) prior to exposure to DOX. Pretreatment with either 400 µM NaHS or 3 µM SB203580, a selective inhibitor of p38 MAPK, ameliorated DOX-induced cardiomyocyte injuries, as evidenced by an increase in cell viability, and decreases in the number of apoptotic cells, ROS generation as well as dissipation of MMP. In conclusion, the findings of the present study demonstrated that the activation of p38 MAPK contributes to DOX-induced injuries, including cytotoxicity, apoptosis, mitochondrial damage and oxidative stress in H9c2 cells. We also provide novel evidence that exogenous H2S protects H9c2 cells against DOX-induced cardiotoxicity by inhibition of the p38 MAPK pathway.
Collapse
Affiliation(s)
- Runmin Guo
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, PR China
| | | | | | | | | | | | | |
Collapse
|
164
|
Lin ST, Chou HC, Chen YW, Chan HL. Redox-proteomic analysis of doxorubicin-induced altered thiol activity in cardiomyocytes. MOLECULAR BIOSYSTEMS 2013; 9:447-56. [DOI: 10.1039/c2mb25367d] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
165
|
Fong MY, Jin S, Rane M, Singh RK, Gupta R, Kakar SS. Withaferin A synergizes the therapeutic effect of doxorubicin through ROS-mediated autophagy in ovarian cancer. PLoS One 2012; 7:e42265. [PMID: 22860102 PMCID: PMC3408484 DOI: 10.1371/journal.pone.0042265] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Accepted: 07/02/2012] [Indexed: 12/27/2022] Open
Abstract
Application of doxorubicin (Dox) for the treatment of cancer is restricted due to its severe side effects. We used combination strategy by combining doxorubicin (Dox) with withaferin A (WFA) to minimize the ill effects of Dox. Treatment of various epithelial ovarian cancer cell lines (A2780, A2780/CP70 and CaOV3) with combination of WFA and Dox (WFA/DOX) showed a time- and dose-dependent synergistic effect on inhibition of cell proliferation and induction of cell death, thus reducing the dosage requirement of Dox. Combination treatment resulted in a significant enhancement of ROS production resulting in immense DNA damage, induction of autophagy analyzed by transmission electron microscope and increase in expression of autophagy marker LC3B, and culminated in cell death analyzed by cleaved caspase 3. We validated combination therapy on tumor growth using an in vitro 3Dimension (3D) tumor model and the more classic in vivo xenograft model of ovarian cancer. Both tumor models showed a 70 to 80% reduction in tumor growth compared to control or animals treated with WFA or Dox alone. Immunohistochemical analysis of the tumor tissues from animals treated with WFA/Dox combination showed a significant reduction in cell proliferation and formation of microvessels accompanied by increased in LC3B level, cleaved caspase 3, and DNA damage. Taken together, our data suggest that combining WFA with Dox decreases the dosage requirement of Dox, therefore, minimizing/eliminating the severe side effects associated with high doses of DOX, suggesting the application of this combination strategy for the treatment of ovarian and other cancers with no or minimum side effects.
Collapse
Affiliation(s)
- Miranda Y. Fong
- Department of Physiology and Biophysics, University of Louisville, Louisville, Kentucky, United States of America
| | - Shunying Jin
- Department of Medicine, University of Louisville, Louisville, Kentucky, United States of America
| | - Madhavi Rane
- Department of Medicine, University of Louisville, Louisville, Kentucky, United States of America
| | - Raj K. Singh
- Vivo Biosciences Inc., Birmingham, Alabama, United States of America
| | - Ramesh Gupta
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, United States of America
- James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, United States of America
| | - Sham S. Kakar
- Department of Physiology and Biophysics, University of Louisville, Louisville, Kentucky, United States of America
- James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, United States of America
- * E-mail:
| |
Collapse
|
166
|
Chen X, Cvetkovic D, Ma CM, Chen L. Quantitative study of focused ultrasound enhanced doxorubicin delivery to prostate tumor in vivo with MRI guidance. Med Phys 2012; 39:2780-6. [PMID: 22559650 DOI: 10.1118/1.4705346] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
PURPOSE The purpose of this study was to investigate the potential of MR-guided pulsed focused ultrasound (pFUS) for the enhancement of drug uptake in prostate tumors in vivo using doxorubicin (Dox). METHODS An antitumor drug Dox, an orthotopic animal prostate tumor model using human prostate cancer, LNCaP cell line, and a clinical FUS treatment system (InSightec ExAblate 2000) with a 1.5T GE MR scanner were used in this study. First, experiments on a tissue mimic phantom to determine the optimal acoustic power and exposure durations with a 10% duty cycle and a 1 Hz pulse rate were performed. The temperature variation was monitored using real-time MR thermometry. Second, tumor-bearing animals were treated with pFUS. There were three groups (n = 8/group): group 1 received pFUS + Dox (10 mg/kg i.v. injection immediately after pFUS exposure), group 2 received Dox only (10 mg/kg i.v. injection), and group 3 was a control. Animals were euthanized 2 h after the pFUS treatment. The Dox concentration in the treated tumors was measured by quantifying fluorescent tracers using a fluorometer. Third, the histological changes of tumors with and without pFUS treatments were evaluated. Finally, experiments were performed to study the spatial drug distribution in tumors after the pFUS treatment, in which two animals received pFUS + Dox, two animals received Dox only, and one animal was used as control. Two hours following the treatment, animals were euthanized and processed. The Dox distribution was determined using a fluorescence microscope. RESULTS Parametric measurements using a tissue phantom showed that the temperature increased with an increasing acoustic power (from 10 to 50 W) or sonication duration (from 10 to 60 s) with a given acoustic frequency of 1 MHz, duty cycle 10%, and pulse rate 1 Hz. A set of ultrasound parameters was identified with which the temperature elevation was less than 5 °C, which was used for nonthermal pFUS sonication. Increased Dox concentration (14.9 ± 2.5 μg/g) was measured in the pFUS-treated group compared to the Dox-only group (9.5 ± 1.6 μg/g), indicating an approximate 60% increase with p = 0.05. The results were consistent with the increased spatial drug distributions by fluorescence imaging. Histological analysis showed increased extravasation in pFUS-treated prostate tumors suggesting increased drug delivery with pFUS. CONCLUSIONS The results showed that pFUS-enhanced drug uptake in prostate tumors was significant. This increased uptake may be due to increased extravasation by pFUS. Optimal pFUS parameters may exist to maximize the drug uptake, and this study using Dox demonstrated a quantitative method for such systematic parametric studies. In addition, this study may provide useful data for the potential application of pFUS-mediated Dox delivery for prostate tumor therapy.
Collapse
Affiliation(s)
- Xiaoming Chen
- Department of Radiation Oncology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | | | | | | |
Collapse
|
167
|
Postescu ID, Chereches G, Tatomir C, Daicoviciu D, Filip GA. Modulation of Doxorubicin-Induced Oxidative Stress by a Grape (Vitis viniferaL.) Seed Extract in Normal and Tumor Cells. J Med Food 2012; 15:639-45. [DOI: 10.1089/jmf.2011.0291] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Ion Dan Postescu
- Department of Radiobiology and Tumor Biology, “Prof. Dr. I. Chiricuta” Oncologic Institute, Cluj-Napoca, Romania
| | - Gabriela Chereches
- Department of Radiobiology and Tumor Biology, “Prof. Dr. I. Chiricuta” Oncologic Institute, Cluj-Napoca, Romania
| | - Corina Tatomir
- Department of Radiobiology and Tumor Biology, “Prof. Dr. I. Chiricuta” Oncologic Institute, Cluj-Napoca, Romania
| | - Doina Daicoviciu
- Department of Physiology, “Iuliu Hatieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Gabriela Adriana Filip
- Department of Physiology, “Iuliu Hatieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
| |
Collapse
|
168
|
Tesei A, Brigliadori G, Carloni S, Fabbri F, Ulivi P, Arienti C, Sparatore A, Del Soldato P, Pasini A, Amadori D, Silvestrini R, Zoli W. Organosulfur derivatives of the HDAC inhibitor valproic acid sensitize human lung cancer cell lines to apoptosis and to cisplatin cytotoxicity. J Cell Physiol 2012; 227:3389-96. [DOI: 10.1002/jcp.24039] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
169
|
Doxorubicin-induced Changes of Ventricular Repolarization Heterogeneity: Results of a Chronic Rat Study. Cardiovasc Toxicol 2012; 12:312-7. [DOI: 10.1007/s12012-012-9172-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
170
|
Chen B, Zhong L, Roush SF, Pentassuglia L, Peng X, Samaras S, Davidson JM, Sawyer DB, Lim CC. Disruption of a GATA4/Ankrd1 signaling axis in cardiomyocytes leads to sarcomere disarray: implications for anthracycline cardiomyopathy. PLoS One 2012; 7:e35743. [PMID: 22532871 PMCID: PMC3332030 DOI: 10.1371/journal.pone.0035743] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2011] [Accepted: 03/23/2012] [Indexed: 01/07/2023] Open
Abstract
Doxorubicin (Adriamycin) is an effective anti-cancer drug, but its clinical usage is limited by a dose-dependent cardiotoxicity characterized by widespread sarcomere disarray and loss of myofilaments. Cardiac ankyrin repeat protein (CARP, ANKRD1) is a transcriptional regulatory protein that is extremely susceptible to doxorubicin; however, the mechanism(s) of doxorubicin-induced CARP depletion and its specific role in cardiomyocytes have not been completely defined. We report that doxorubicin treatment in cardiomyocytes resulted in inhibition of CARP transcription, depletion of CARP protein levels, inhibition of myofilament gene transcription, and marked sarcomere disarray. Knockdown of CARP with small interfering RNA (siRNA) similarly inhibited myofilament gene transcription and disrupted cardiomyocyte sarcomere structure. Adenoviral overexpression of CARP, however, was unable to rescue the doxorubicin-induced sarcomere disarray phenotype. Doxorubicin also induced depletion of the cardiac transcription factor GATA4 in cardiomyocytes. CARP expression is regulated in part by GATA4, prompting us to examine the relationship between GATA4 and CARP in cardiomyocytes. We show in co-transfection experiments that GATA4 operates upstream of CARP by activating the proximal CARP promoter. GATA4-siRNA knockdown in cardiomyocytes inhibited CARP expression and myofilament gene transcription, and induced extensive sarcomere disarray. Adenoviral overexpression of GATA4 (AdV-GATA4) in cardiomyocytes prior to doxorubicin exposure maintained GATA4 levels, modestly restored CARP levels, and attenuated sarcomere disarray. Interestingly, siRNA-mediated depletion of CARP completely abolished the Adv-GATA4 rescue of the doxorubicin-induced sarcomere phenotype. These data demonstrate co-dependent roles for GATA4 and CARP in regulating sarcomere gene expression and maintaining sarcomeric organization in cardiomyocytes in culture. The data further suggests that concurrent depletion of GATA4 and CARP in cardiomyocytes by doxorubicin contributes in large part to myofibrillar disarray and the overall pathophysiology of anthracycline cardiomyopathy.
Collapse
Affiliation(s)
- Billy Chen
- Molecular Medicine Program, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Lin Zhong
- Division of Cardiovascular Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Sarah F. Roush
- Division of Cardiovascular Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Laura Pentassuglia
- Division of Cardiovascular Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Xuyang Peng
- Division of Cardiovascular Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Susan Samaras
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Jeffrey M. Davidson
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
- Research Service, Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee, United Sates of America
| | - Douglas B. Sawyer
- Division of Cardiovascular Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Chee Chew Lim
- Division of Cardiovascular Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
- * E-mail:
| |
Collapse
|
171
|
Kharin S, Krandycheva V, Tsvetkova A, Strelkova M, Shmakov D. Remodeling of ventricular repolarization in a chronic doxorubicin cardiotoxicity rat model. Fundam Clin Pharmacol 2012; 27:364-72. [DOI: 10.1111/j.1472-8206.2012.01037.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
172
|
Colak MC, Parlakpinar H, Tasdemir S, Samdanci E, Kose E, Polat A, Sarihan E, Acet A. Therapeutic effects of ivabradine on hemodynamic parameters and cardiotoxicity induced by doxorubicin treatment in rat. Hum Exp Toxicol 2012; 31:945-54. [DOI: 10.1177/0960327112438288] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The aim of this study was to investigate the possible effects of ivabradine against doxorubicin (DOX)-induced cardiotoxicity in rats using hemodynamic parameters (electrocardiogram, heart rate (HR), and blood pressure), biochemical markers of oxidative stress, lactate dehydrogenase, aspartate transaminase, creatine kinase-MB, and histopathological analyses both in serum and tissue specimens. A total of 28 female rats were randomly assigned to 4 groups: (a) control ( n = 6 rats), (b) DOX group ( n = 7 rats), (c) DOX + ivabradine–treated group ( n = 8 rats), and (d) ivabradine group ( n = 7 rats). When the means of the four groups were compared, there was only a significant difference in the level of HR ( p < 0.05). DOX treatment caused more HR elevation when compared to the control group, whereas ivabradine application after DOX treatment significantly reduced HR levels. Cardiomyocytes were revealed as normal histology in the light of both hematoxylin and eosin staining and immunostaining methods (caspase-3 and bcl-2) in all groups. The present study reported the therapeutic effects of ivabradine against DOX-induced cardiotoxicity accompanied by the hemodynamic and biochemical parameters.
Collapse
Affiliation(s)
- MC Colak
- Department of Cardiovascular Surgery, Inonu University, Malatya, Turkey
| | - H Parlakpinar
- Department of Pharmacology, Inonu University, Malatya, Turkey
| | - S Tasdemir
- Department of Pharmacology, Inonu University, Malatya, Turkey
| | - E Samdanci
- Department of Pathology, Inonu University, Malatya, Turkey
| | - E Kose
- Department of Anatomy, Inonu University, Malatya, Turkey
| | - A Polat
- Department of Physiology, Inonu University, Malatya, Turkey
| | - E Sarihan
- Department of Emergency Medicine, Inonu University, Malatya, Turkey
| | - A Acet
- Department of Pharmacology, Inonu University, Malatya, Turkey
| |
Collapse
|
173
|
Redfors B, Shao Y, Råmunddal T, Lindbom M, Täng MS, Stillemark-Billton P, Boren J, Omerovic E. Effects of doxorubicin on myocardial expression of apolipoprotein-B. SCAND CARDIOVASC J 2012; 46:93-8. [PMID: 22263831 DOI: 10.3109/14017431.2012.653825] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE Doxorubicin (DOX) is an effective antitumour agent against a variety of human malignancies but is associated with deleterious side effects, including myocardial damage and heart failure. Myocardial apoB-containing lipoprotein (apoB) is upregulated post myocardial infarction and has been shown to be cardioprotective in this setting by unloading excessive lipid. The aim of this study was to investigate whether apoB expression is increased also in DOX-induced heart failure and whether apoB overexpression protects the heart in DOX-induced myocardial injury. DESIGN Cardiac function and energy metabolism was studied in mice and rats 24 hours after intraperitoneally administered DOX. RESULTS We found that the content of apoB was decreased in rat myocardium 24 hours after DOX injection. In contrast, apoB content was increased in the infarcted myocardium of rats 24 hours post ischemia-reperfusion. Moreover, transgenic mice overexpressing apoB had better cardiac function and lower intracellular lipid accumulation compared to wild type mice 24 hours post DOX. CONCLUSIONS Our findings indicate that depression of the myocardial apoB system may contribute to DOX-induced cardiac injury and that overexpression of apoB is protective, not only in ischemically damaged myocardium, but also in DOX-induced heart failure.
Collapse
Affiliation(s)
- Bjorn Redfors
- The Wallenberg laboratory at Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|
174
|
Redfors B, Shao Y, Råmunddal T, Lindbom M, Täng MS, Stillemark-Billton P, Boren J, Omerovic E. Effects of doxorubicin on myocardial expression of apolipoprotein-B. SCAND CARDIOVASC J 2011. [DOI: 10.3109/14017431.2011.653825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
175
|
Cationic albumin nanoparticles for enhanced drug delivery to treat breast cancer: preparation and in vitro assessment. JOURNAL OF DRUG DELIVERY 2011; 2012:686108. [PMID: 22187654 PMCID: PMC3236506 DOI: 10.1155/2012/686108] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Accepted: 09/10/2011] [Indexed: 11/20/2022]
Abstract
Most anticancer drugs are greatly limited by the serious side effects that they cause. Doxorubicin (DOX) is an antineoplastic agent, commonly used against breast cancer. However, it may lead to irreversible cardiotoxicity, which could even result in congestive heart failure. In order to avoid these harmful side effects to the patients and to improve the therapeutic efficacy of doxorubicin, we developed DOX-loaded polyethylenimine- (PEI-) enhanced human serum albumin (HSA) nanoparticles. The formed nanoparticles were ~137 nm in size with a surface zeta potential of ~+15 mV, prepared using 20 μg of PEI added per mg of HSA. Cytotoxicity was not observed with empty PEI-enhanced HSA nanoparticles, formed with low-molecular weight (25 kDa) PEI, indicating biocompatibility and safety of the nanoparticle formulation. Under optimized transfection conditions, approximately 80% of cells were transfected with HSA nanoparticles containing tetramethylrhodamine-conjugated bovine serum albumin. Conclusively, PEI-enhanced HSA nanoparticles show potential for developing into an effective carrier for anticancer drugs.
Collapse
|
176
|
Zong WN, Yang XH, Chen XM, Huang HJ, Zheng HJ, Qin XY, Yong YH, Cao K, Huang J, Lu XZ. Regulation of angiotensin-(1-7) and angiotensin II type 1 receptor by telmisartan and losartan in adriamycin-induced rat heart failure. Acta Pharmacol Sin 2011; 32:1345-50. [PMID: 21963897 DOI: 10.1038/aps.2011.96] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
AIM To investigate the possible effects of telmisartan and losartan on cardiac function in adriamycin (ADR)-induced heart failure in rats, and to explore the changes in plasma level of angiotensin-(1-7)[Ang-(1-7)] and myocardial expression of angiotensin II type 1/2 receptors (AT(1)R / AT(2)R) and Mas receptor caused by the two drugs. METHODS Male Sprague-Dawley rats were randomly divided into 4 groups: the control group, ADR-treated heart failure group (ADR-HF), telmisartan plus ADR-treated group (Tel+ADR) and losartan plus ADR-treated group (Los+ADR). ADR was administrated (2.5 mg/kg, ip, 6 times in 2 weeks). The rats in the Tel+ADR and Los+ADR groups were treated orally with telmisartan (10 mg/kg daily po) and losartan (30 mg/kg daily), respectively, for 6 weeks. The plasma level of Ang-(1-7) was determined using ELISA. The mRNA and protein expression of myocardial Mas receptor, AT(1)R and AT(2)R were measured using RT-PCR and Western blotting, respectively. RESULTS ADR significantly reduced the plasma level of Ang-(1-7) and the expression of myocardial Mas receptor and myocardial AT(2)R, while significantly increased the expression of myocardial AT(1)R. Treatment with telmisartan and losartan effectively increased the plasma level of Ang-(1-7) and suppressed myocardial AT(1)R expression, but did not influence the expression of Mas receptor and AT(2)R. CONCLUSION The protective effects of telmisartan and losartan in ADR-induced heart failure may be partially due to regulation of circulating Ang-(1-7) and myocardial AT(1)R expression.
Collapse
|
177
|
Propofol ameliorates doxorubicin-induced oxidative stress and cellular apoptosis in rat cardiomyocytes. Toxicol Appl Pharmacol 2011; 257:437-48. [PMID: 22015447 DOI: 10.1016/j.taap.2011.10.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2011] [Revised: 09/18/2011] [Accepted: 10/03/2011] [Indexed: 12/15/2022]
Abstract
BACKGROUND Propofol is an anesthetic with pluripotent cytoprotective properties against various extrinsic insults. This study was designed to examine whether this agent could also ameliorate the infamous toxicity of doxorubicin, a widely-used chemotherapeutic agent against a variety of cancer diseases, on myocardial cells. METHODS Cultured neonatal rat cardiomyocytes were administrated with vehicle, doxorubicin (1μM), propofol (1μM), or propofol plus doxorubicin (given 1h post propofol). After 24h, cells were harvested and specific analyses regarding oxidative/nitrative stress and cellular apoptosis were conducted. RESULTS Trypan blue exclusion and MTT assays disclosed that viability of cardiomyocytes was significantly reduced by doxorubicin. Contents of reactive oxygen and nitrogen species were increased and antioxidant enzymes SOD1, SOD2, and GPx were decreased in these doxorubicin-treated cells. Mitochondrial dehydrogenase activity and membrane potential were also depressed, along with activation of key effectors downstream of mitochondrion-dependent apoptotic signaling. Besides, abundance of p53 was elevated and cleavage of PKC-δ was induced in these myocardial cells. In contrast, all of the above oxidative, nitrative and pro-apoptotic events could be suppressed by propofol pretreatment. CONCLUSIONS Propofol could extensively counteract oxidative/nitrative and multiple apoptotic effects of doxorubicin in the heart; hence, this anesthetic may serve as an adjuvant agent to assuage the untoward cardiac effects of doxorubicin in clinical application.
Collapse
|
178
|
Chang WT, Li J, Haung HH, Liu H, Han M, Ramachandran S, Li CQ, Sharp WW, Hamann KJ, Yuan CS, Hoek TLV, Shao ZH. Baicalein protects against doxorubicin-induced cardiotoxicity by attenuation of mitochondrial oxidant injury and JNK activation. J Cell Biochem 2011; 112:2873-81. [PMID: 21618589 PMCID: PMC3178681 DOI: 10.1002/jcb.23201] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The cardiotoxicity of doxorubicin limits its clinical use in the treatment of a variety of malignancies. Previous studies suggest that doxorubicin-associated cardiotoxicity is mediated by reactive oxygen species (ROS)-induced apoptosis. We therefore investigated if baicalein, a natural antioxidant component of Scutellaria baicalensis, could attenuate ROS generation and cell death induced by doxorubicin. Using an established chick cardiomyocyte model, doxorubicin (10 µM) increased cell death in a concentration- and time-dependent manner. ROS generation was increased in a dose-response fashion and associated with loss of mitochondrial membrane potential. Doxorubicin also augmented DNA fragmentation and increased the phosphorylation of ROS-sensitive pro-apoptotic kinase c-Jun N-terminal kinase (JNK). Adjunct treatment of baicalein (25 µM) and doxorubicin for 24 h significantly reduced both ROS generation (587 ± 89 a.u. vs. 932 a.u. ± 121 a.u., P < 0.01) and cell death (30.6 ± 5.1% vs. 46.8 ± 8.3%, P < 0.01). The dissipated mitochondrial potential and increased DNA fragmentation were also ameliorated. Along with the reduction of ROS and apoptosis, baicalein attenuated phosphorylation of JNK induced by doxorubicin (1.7 ± 0.3 vs. 3.0 ± 0.4-fold, P < 0.05). Co-treatment of cardiomyocytes with doxorubicin and JNK inhibitor SP600125 (10 µM; 24 h) reduced JNK phosphorylation and enhanced cell survival, suggesting that the baicalein protection against doxorubicin cardiotoxicity was mediated by JNK activation. Importantly, concurrent baicalein treatment did not interfere with the anti-proliferative effects of doxorubicin in human breast cancer MCF-7 cells. In conclusion, baicalein adjunct treatment confers anti-apoptotic protection against doxorubicin-induced cardiotoxicity without compromising its anti-cancer efficacy.
Collapse
Affiliation(s)
- Wei-Tien Chang
- Emergency Resuscitation Center, Section of Emergency Medicine, Department of Medicine, University of Chicago, Chicago, Illinois 60637, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
179
|
Zhang Y, Xiang J, Liu Y, Zhang X, Tang Y. Constructing transferrin receptor targeted drug delivery system by using doxorubicin hydrochloride and vanadocene dichloride. Bioorg Med Chem Lett 2011; 21:5982-6. [DOI: 10.1016/j.bmcl.2011.07.066] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2011] [Revised: 06/30/2011] [Accepted: 07/14/2011] [Indexed: 10/17/2022]
|
180
|
Ajith TA, Janardhanan KK. Antimutagenic effect of Phellinus rimosus (Berk) Pilat against chemical induced mutations of histidine dependent Salmonella typhimurium strains. Food Chem Toxicol 2011; 49:2676-2680. [PMID: 21784121 DOI: 10.1016/j.fct.2011.07.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2011] [Revised: 07/07/2011] [Accepted: 07/08/2011] [Indexed: 10/18/2022]
Abstract
Mutations are one of the important factors contributing to oncogenesis. Somatic mutations have been detected in oncogenes and tumor suppressor genes in various types of cancers. In vitro antimutagenic activity of ethyl acetate extract of macro fungus, Phellinus rimosus was evaluated by Ames' mutagenicity assay. The effect was evaluated against the direct acting mutagens (sodium azide, N-methyl-N'-nitro-N-nitrosoguanidine, doxorubicin and 4-nitro-o-phenylenediamine) and mutagen needing activation (2-acetyl aminofluorine, and benzo[a]pyrene). The extract was significantly (p<0.05) and dose dependently effective against direct acting mutagens and mutagen needing activation. Among the antimutagenic activity against directly acting mutagens, effect was found to be highest against doxorubicin-induced mutation. The antimutagenic effect of the extract against indirect acting mutagen in the presence of mammalian metabolic activation system was also found to be significant (p<0.01). The background bacterial growth and number of revertant colonies in the extract alone treated plate with or with out metabolic activator was almost same as that of spontaneous revertants. This indicated the non-toxic nature of the extract. The effect was partially ascribed to the antioxidant activity. The results of the study suggest the possible antitumor mechanisms of P. rimosus.
Collapse
Affiliation(s)
- T A Ajith
- Department of Biochemistry, Amala Institute of Medical Sciences, Amala Nagar, Thrissur 680 555, Kerala, India
| | | |
Collapse
|
181
|
Stoodley PW, Richards DAB, Hui R, Boyd A, Harnett PR, Meikle SR, Clarke J, Thomas L. Two-dimensional myocardial strain imaging detects changes in left ventricular systolic function immediately after anthracycline chemotherapy. EUROPEAN JOURNAL OF ECHOCARDIOGRAPHY 2011; 12:945-52. [DOI: 10.1093/ejechocard/jer187] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
182
|
Brechbuhl HM, Kachadourian R, Min E, Chan D, Day BJ. Chrysin enhances doxorubicin-induced cytotoxicity in human lung epithelial cancer cell lines: the role of glutathione. Toxicol Appl Pharmacol 2011; 258:1-9. [PMID: 21856323 DOI: 10.1016/j.taap.2011.08.004] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2011] [Revised: 07/28/2011] [Accepted: 08/02/2011] [Indexed: 01/28/2023]
Abstract
We hypothesized that flavonoid-induced glutathione (GSH) efflux through multi-drug resistance proteins (MRPs) and subsequent intracellular GSH depletion is a viable mechanism to sensitize cancer cells to chemotherapies. This concept was demonstrated using chrysin (5-25 μM) induced GSH efflux in human non-small cell lung cancer lines exposed to the chemotherapeutic agent, doxorubicin (DOX). Treatment with chrysin resulted in significant and sustained intracellular GSH depletion and the GSH enzyme network in the four cancer cell types was predictive of the severity of chrysin induced intracellular GSH depletion. Gene expression data indicated a positive correlation between basal MRP1, MRP3 and MRP5 expression and total GSH efflux before and after chrysin exposure. Co-treating the cells for 72 h with chrysin (5-30 μM) and DOX (0.025-3.0 μM) significantly enhanced the sensitivity of the cells to DOX as compared to 72-hour DOX alone treatment in all four cell lines. The maximum decrease in the IC(50) values of cells treated with DOX alone compared to co-treatment with chrysin and DOX was 43% in A549 cells, 47% in H157 and H1975 cells and 78% in H460 cells. Chrysin worked synergistically with DOX to induce cancer cell death. This approach could allow for use of lower concentrations and/or sensitize cancer cells to drugs that are typically resistant to therapy.
Collapse
|
183
|
Smuder AJ, Kavazis AN, Min K, Powers SK. Exercise protects against doxorubicin-induced markers of autophagy signaling in skeletal muscle. J Appl Physiol (1985) 2011; 111:1190-8. [PMID: 21778418 DOI: 10.1152/japplphysiol.00429.2011] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Doxorubicin (DOX) is an effective antitumor agent used in cancer treatment. Unfortunately, DOX is also toxic to skeletal muscle and can result in significant muscle wasting. The cellular mechanism(s) by which DOX induces toxicity in skeletal muscle fibers remains unclear. Nonetheless, DOX-induced toxicity is associated with increased generation of reactive oxygen species, oxidative damage, and activation of the calpain and caspase-3 proteolytic systems within muscle fibers. It is currently unknown if autophagy, a proteolytic system that can be triggered by oxidative stress, is activated in skeletal muscles following DOX treatment. Therefore, we tested the hypothesis that systemic administration of DOX leads to increased expression of autophagy markers in the rat soleus muscle. Our results reveal that DOX administration results in increased muscle mRNA levels and/or protein abundance of several important autophagy proteins, including: Beclin-1, Atg12, Atg7, LC3, LC3II-to-LCI ratio, and cathepsin L. Furthermore, given that endurance exercise increases skeletal muscle antioxidant capacity and protects muscle against DOX-induced oxidative stress, we performed additional experiments to determine whether exercise training before DOX administration would attenuate DOX-induced increases in expression of autophagy genes. Our results clearly show that exercise can protect skeletal muscle from DOX-induced expression of autophagy genes. Collectively, our findings indicate that DOX administration increases the expression of autophagy genes in skeletal muscle, and that exercise can protect skeletal muscle against DOX-induced activation of autophagy.
Collapse
Affiliation(s)
- Ashley J Smuder
- Department of Applied Physiology and Kinesiology, Center for Exercise Science, University of Florida, Gainesville, Florida, USA
| | | | | | | |
Collapse
|
184
|
Zhang Y, Kang YM, Tian C, Zeng Y, Jia LX, Ma X, Du J, Li HH. Overexpression of Nrdp1 in the heart exacerbates doxorubicin-induced cardiac dysfunction in mice. PLoS One 2011; 6:e21104. [PMID: 21738612 PMCID: PMC3124482 DOI: 10.1371/journal.pone.0021104] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2011] [Accepted: 05/19/2011] [Indexed: 12/01/2022] Open
Abstract
Background Cardiac cell death and generation of oxidative stress contribute to doxorubicin (DOX)-induced cardiac dysfunction. E3 ligase Nrdp1 plays a critical role in the regulation of cell apoptosis, inflammation and production of reactive oxygen species (ROS), which may contribute to heart failure. However, the role of Nrdp1 in DOX-induced cardiac injury remains to be determined. Methods and Results We examined the effect of Nrdp1 overexpression with DOX treatment in rat neonatal cardiomyocytes and mouse heart tissue. Cardiomyocytes were infected with adenovirus containing GFP (Ad-GFP), Nrdp1 wild-type (Ad-Nrdp1) or the dominant-negative form of Nrdp1 (Ad-Dn-Nrdp1), then treated with DOX for 24 hr. DOX treatment increased cell death and apoptosis, with Ad-Nrdp1 infection enhancing these actions but Ad-Dn-Nrdp1 infection attenuating these effects. Furthermore, 5 days after a single injection of DOX (20 mg/kg, intraperitoneally), Nrdp1 transgenic mice (TG) showed decreased cardiac function and increased apoptosis, autophagy and oxidative stress as compared with wild-type (WT) mice (P<0.01). Survival rate was significantly lower in Nrdp1 TG mice than in WT mice 10 days after DOX injection (P<0.01). Conclusions/Significance These results were associated with decreased activation of Akt, extracellular signal-regulated kinase 1/2 (ERK1/2) and signal transducer and activator of transcription 3 (STAT3) signaling pathways. Nrdp1 may be a key mediator in the development of cardiac dysfunction after DOX treatment and associated with inhibition of Akt, ERK1/2 and STAT3. Nrdp1 may be a new therapeutic target in protecting against the cardiotoxic effects of DOX.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Pathology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | | | | | | | | | | | | | | |
Collapse
|
185
|
Sauter KAD, Wood LJ, Wong J, Iordanov M, Magun BE. Doxorubicin and daunorubicin induce processing and release of interleukin-1β through activation of the NLRP3 inflammasome. Cancer Biol Ther 2011; 11:1008-16. [PMID: 21464611 PMCID: PMC3142364 DOI: 10.4161/cbt.11.12.15540] [Citation(s) in RCA: 158] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Revised: 03/09/2011] [Accepted: 03/17/2011] [Indexed: 01/21/2023] Open
Abstract
Anthracyclines including doxorubicin and daunorubicin are commonly used for the treatment of both hematologic and solid tumors. Dose related adverse effects often limit the effectiveness of anthracyclines in chemotherapy. Drug-related systemic inflammation mediated by interleukin-1beta (IL-1β) has been implicated in contributing to these adverse effects. The molecular mechanisms underlying anthracycline-mediated expression and IL-1β release are not understood. Elucidating the molecular basis by which anthracyclines upregulate IL-1β activity may present opportunities to decrease the inflammatory consequences of these drugs. Here we demonstrate that doxorubicin induces a systemic increase in IL-1β and other inflammatory cytokines, chemokines and growth factors including TNF-α, IL-6, CXCL1/Gro-α, CCL2/MCP-1, granulocyte colony stimulating factor (GCSF), and CXCL10/IP-10. Studies with IL-1R-deficient mice demonstrate that IL-1 signaling plays a role in doxorubicin-induced increases in IL-6 and GCSF. In vitro studies with doxorubicin and daunorubicin failed to induce expression of proIL-1β in unprimed murine bone marrow-derived macrophages (BMDM) but enhanced the expression of proIL-1β in BMDM that had previously been primed with LPS. Furthermore, doxorubicin and daunorubicin induced the processing and release of IL-1β from LPS-primed BMDM by providing danger signals that lead to assembly and activation of the inflammasome. The release of IL-1β required the expression of ASC, caspase-1, and NLRP3, demonstrating that doxorubicin and daunorubicin-induced inflammation is mediated by the NLRP3 inflammasome. As with other agents that induce activation of the NLRP3 inflammasome, the ability of doxorubicin to provide proinflammatory danger signals was inhibited by co-treatment of cells with ROS inhibitors or by incubating cells in high extracellular potassium. These studies suggest that proinflammatory responses to anthracycline chemotherapeutic agents are mediated, at least in part, by promoting the processing and release of IL-1β, and that some of the adverse inflammatory consequences that complicate chemotherapy with anthracyclines may be reduced by suppressing the actions of IL-1β.
Collapse
Affiliation(s)
- Kristin A D Sauter
- Department of Cell and Developmental Biology, Oregon Health and Science University, Portland, USA
| | | | | | | | | |
Collapse
|
186
|
Yang F, Chen Z, Pan J, Li X, Feng J, Yang H. An integrated microfluidic array system for evaluating toxicity and teratogenicity of drugs on embryonic zebrafish developmental dynamics. BIOMICROFLUIDICS 2011; 5:24115. [PMID: 21799721 PMCID: PMC3145240 DOI: 10.1063/1.3605509] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2011] [Accepted: 06/08/2011] [Indexed: 05/03/2023]
Abstract
Seeking potential toxic and side effects for clinically available drugs is considerably beneficial in pharmaceutical safety evaluation. In this article, the authors developed an integrated microfluidic array system for phenotype-based evaluation of toxic and teratogenic potentials of clinical drugs by using zebrafish (Danio rerio) embryos as organism models. The microfluidic chip consists of a concentration gradient generator from upstream and an array of open embryonic culture structures by offering continuous stimulation in gradients and providing guiding, cultivation and exposure to the embryos, respectively. The open culture reservoirs are amenable to long-term embryonic culturing. Gradient test substances were delivered in a continuous or a developmental stage-specific manner, to induce embryos to generate dynamic developmental toxicity and teratogenicity. Developmental toxicity of doxorubicin on zebrafish eggs were quantitatively assessed via heart rate, and teratological effects were characterized by pericardial impairment, tail fin, notochord, and SV-BA distance ∕body length. By scoring the teratogenic severity, we precisely evaluated the time- and dose-dependent damage on the chemical-exposed embryos. The simple and easily operated method presented herein demonstrates that zebrafish embryo-based pharmaceutic assessment could be performed using microfluidic systems and holds a great potential in high-throughput screening for new compounds at single animal resolution.
Collapse
Affiliation(s)
- Fan Yang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, China
| | | | | | | | | | | |
Collapse
|
187
|
Li L, Zhao X, Yang C, Hu H, Qiao M, Chen D. Preparation and optimization of doxorubicin-loaded albumin nanoparticles using response surface methodology. Drug Dev Ind Pharm 2011; 37:1170-80. [PMID: 21449822 DOI: 10.3109/03639045.2011.563781] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND The objective of this work was to optimize the preparation of doxorubicin-loaded albumin nanoparticles (Dox-A-Nps) through desolvation procedures using response surface methodology (RSM). A central composite design (CCD) for four factors at five levels was used in this study. METHOD Albumin nanoparticles were prepared through a desolvation method and were optimized in the aid of CCD. Albumin concentration, amount of doxorubicin, pH values, and percentage of glutaraldehyde were selected as independent variables, particle size, zeta potential, drug loading, encapsulation efficiency, and nanoparticles yield were chosen as response variables. RSM and multiple response optimizations utilizing a quadratic polynomial equation were used to obtain an optimal formulation. RESULTS The optimal formulation for Dox-A-Nps was composed of albumin concentration of 17 mg/ml, amount of doxorubicin of 2 mg/ml, pH value is 9 and percentage of glutaraldehyde of 125% of the theoretic amount, under which the optimized conditions gave rise to the actual average value of mean particle size (151 ± 0.43 nm), zeta potential (-18.8 ± 0.21 mV), drug loading efficiency (21.4 ± 0.70%), drug entrapment efficiency (76.9 ± 0.21%) and nanoparticles yield (82.0 ± 0.34%). The storage stability experiments proved that Dox-A-Nps stable in 4°C over the period of 4 months. The in vitro experiments showed a burst release at the initial stage and followed by a prolonged release of Dox from albumin nanoparticles up to 60 h. CONCLUSIONS This study showed that the RSM-CCD method could efficiently be applied for the modeling of nanoparticles, which laid the foundation of the further research of immuno nanoparticles.
Collapse
Affiliation(s)
- Lei Li
- School of Pharmacy, Shenyang Pharmaceutical University, PR China
| | | | | | | | | | | |
Collapse
|
188
|
The mitochondrial permeability transition pore and the cardiac necrotic program. Pediatr Cardiol 2011; 32:258-62. [PMID: 21210090 DOI: 10.1007/s00246-010-9880-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Accepted: 12/17/2010] [Indexed: 12/27/2022]
Abstract
That apoptosis is mediated by specific pathways has long been established. However, more recent data have begun to suggest that necrosis may in fact be "programmed" and not a default "accidental" pathway as previously thought. The mitochondrial permeability transition pore, a known contributor to the development of many cardiac diseases, is emerging as one among several mediators of this necrotic program. Consequently, this report briefly reviews the roles of necrosis versus apoptosis in the pathogenesis of cardiac disease and discusses the role that the mitochondrial pore plays in cardiac necrotic cell death.
Collapse
|
189
|
Tangpong J, Miriyala S, Noel T, Sinthupibulyakit C, Jungsuwadee P, St. Clair DK. Doxorubicin-induced central nervous system toxicity and protection by xanthone derivative of Garcinia mangostana. Neuroscience 2011; 175:292-9. [PMID: 21074598 PMCID: PMC3136166 DOI: 10.1016/j.neuroscience.2010.11.007] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2010] [Revised: 10/29/2010] [Accepted: 11/05/2010] [Indexed: 12/20/2022]
Abstract
Doxorubicin (Dox) is a potent, broad-spectrum chemotherapeutic drug used around the world. Despite its effectiveness, it has a wide range of toxic side effects, many of which most likely result from its inherent pro-oxidant activity. It has been reported that Dox has toxic effects on normal tissues, including brain tissue. The present study tested the protective effect of a xanthone derivative of Garcinia Mangostana against Dox-induced neuronal toxicity. Xanthone can prevent Dox from causing mononuclear cells to increase the level of tumor necrosis factor-alpha (TNFα). We show that xanthone given to mice before Dox administration suppresses protein carbonyl, nitrotyrosine and 4-hydroxy-2'-nonenal (4HNE)-adducted proteins in brain tissue. The levels of the pro-apoptotic proteins p53 and Bax and the anti-apoptotic protein Bcl-xL were significantly increased in Dox-treated mice compared with the control group. Consistent with the increase of apoptotic markers, the levels of caspase-3 activity and TUNEL-positive cells were also increased in Dox-treated mice. Pretreatment with xanthone suppressed Dox-induced increases in all indicators of injury tested. Together, the results suggest that xanthone prevents Dox-induced central nervous system toxicity, at least in part, by suppression of Dox-mediated increases in circulating TNFα. Thus, xanthone is a good candidate for prevention of systemic effects resulting from reactive oxygen generating anticancer therapeutics.
Collapse
Affiliation(s)
- J. Tangpong
- School of Allied Health Sciences and Public Health, 222 Thaiburi, Thasala, Nakhon-Si-Thammarat, 80160, Thailand
| | - S. Miriyala
- Graduate Center for Toxicology, 1095 VA Drive, 306 Health Sciences Research Building, Lexington, KY 40536-0305, USA
| | - T. Noel
- Graduate Center for Toxicology, 1095 VA Drive, 306 Health Sciences Research Building, Lexington, KY 40536-0305, USA
| | - C. Sinthupibulyakit
- Graduate Center for Toxicology, 1095 VA Drive, 306 Health Sciences Research Building, Lexington, KY 40536-0305, USA
| | - P. Jungsuwadee
- Graduate Center for Toxicology, 1095 VA Drive, 306 Health Sciences Research Building, Lexington, KY 40536-0305, USA
| | - D. K. St. Clair
- Graduate Center for Toxicology, 1095 VA Drive, 306 Health Sciences Research Building, Lexington, KY 40536-0305, USA
| |
Collapse
|
190
|
Dursun N, Taşkın E, Yerer Aycan MB, Şahin L. Selenium-mediated cardioprotection against adriamycin-induced mitochondrial damage. Drug Chem Toxicol 2011; 34:199-207. [DOI: 10.3109/01480545.2010.538693] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
191
|
Smuder AJ, Kavazis AN, Min K, Powers SK. Exercise protects against doxorubicin-induced oxidative stress and proteolysis in skeletal muscle. J Appl Physiol (1985) 2011; 110:935-42. [PMID: 21310889 DOI: 10.1152/japplphysiol.00677.2010] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Doxorubicin (Dox) is a potent antitumor agent used in cancer treatment. Unfortunately, Dox is myotoxic and results in significant reductions in skeletal muscle mass and function. Complete knowledge of the mechanism(s) by which Dox induces toxicity in skeletal muscle is incomplete, but it is established that Dox-induced toxicity is associated with increased generation of reactive oxygen species and oxidative damage within muscle fibers. Since muscular exercise promotes the expression of numerous cytoprotective proteins (e.g., antioxidant enzymes, heat shock protein 72), we hypothesized that muscular exercise will attenuate Dox-induced damage in exercise-trained muscle fibers. To test this postulate, Sprague-Dawley rats were randomly assigned to the following groups: sedentary, exercise, sedentary with Dox, or exercise with Dox. Our results show increased oxidative stress and activation of cellular proteases (calpain and caspase-3) in skeletal muscle of animals treated with Dox. Importantly, our findings reveal that exercise can prevent the Dox-induced oxidative damage and protease activation in the trained muscle. This exercise-induced protection against Dox-induced toxicity may be due, at least in part, to an exercise-induced increase in muscle levels of antioxidant enzymes and heat shock protein 72. Together, these novel results demonstrate that muscular exercise is a useful countermeasure that can protect skeletal muscle against Dox treatment-induced oxidative stress and protease activation in skeletal muscles.
Collapse
Affiliation(s)
- Ashley J Smuder
- Dept. of Applied Physiology and Kinesiology, Univ. of Florida, Gainesville, FL 32611, USA.
| | | | | | | |
Collapse
|
192
|
Zheng J, Lee HCM, bin Sattar MM, Huang Y, Bian JS. Cardioprotective effects of epigallocatechin-3-gallate against doxorubicin-induced cardiomyocyte injury. Eur J Pharmacol 2011; 652:82-8. [DOI: 10.1016/j.ejphar.2010.10.082] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2010] [Revised: 10/06/2010] [Accepted: 10/29/2010] [Indexed: 11/25/2022]
|
193
|
Cytotoxicity and topoisomerase I/II inhibition of glycosylated 2-phenyl-indoles, 2-phenyl-benzo[b]thiophenes and 2-phenyl-benzo[b]furans. Bioorg Med Chem 2011; 19:603-12. [DOI: 10.1016/j.bmc.2010.10.054] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2010] [Revised: 10/21/2010] [Accepted: 10/26/2010] [Indexed: 11/21/2022]
|
194
|
Virgin olive oil minor components as natural drugs for the treatment of breast cancer: preliminary experiments on squalene. MEDITERRANEAN JOURNAL OF NUTRITION AND METABOLISM 2010. [DOI: 10.1007/s12349-010-0019-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
195
|
Konishi M, Haraguchi G, Ohigashi H, Ishihara T, Saito K, Nakano Y, Isobe M. Adiponectin protects against doxorubicin-induced cardiomyopathy by anti-apoptotic effects through AMPK up-regulation. Cardiovasc Res 2010; 89:309-19. [DOI: 10.1093/cvr/cvq335] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
196
|
Stoodley PW, Richards DAB, Meikle SR, Clarke J, Hui R, Thomas L. The potential role of echocardiographic strain imaging for evaluating cardiotoxicity due to cancer therapy. Heart Lung Circ 2010; 20:3-9. [PMID: 20965783 DOI: 10.1016/j.hlc.2010.09.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2010] [Revised: 08/28/2010] [Accepted: 09/06/2010] [Indexed: 01/21/2023]
Abstract
Refinements to conventional treatment and the development of new therapies have led to significant improvements in cancer survival. Yet, many frontline cancer treatments continue to be hindered by their significant side effects, amongst which cardiotoxicity is particularly important. Therefore, the focus of cancer management has changed; treatment is no longer aimed solely at overcoming malignancy, but emphasises early identification and treatment of potential side effects. In this regard, the cardiotoxic potential of certain anticancer agents mandate close monitoring of cardiac function, and the method of choice for monitoring is transthoracic echocardiography. Whilst this method has its limitations, a newer echocardiographic technique called myocardial strain imaging has the potential to detect early sub-clinical changes in cardiac function due to cardiotoxicity. Strain analysis has been the subject of several recent studies to evaluate its potential in monitoring cardiotoxicity, and this article reviews the recent literature and explores the potential role of myocardial strain imaging in cancer management and avenues for future research.
Collapse
Affiliation(s)
- Paul W Stoodley
- Discipline of Medical Radiation Sciences, Faculty of Health Sciences, University of Sydney, NSW, Australia
| | | | | | | | | | | |
Collapse
|
197
|
Morrissy S, Xu B, Aguilar D, Zhang J, Chen QM. Inhibition of apoptosis by progesterone in cardiomyocytes. Aging Cell 2010; 9:799-809. [PMID: 20726854 DOI: 10.1111/j.1474-9726.2010.00619.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
While gender-based differences in heart disease have raised the possibility that estrogen (ES) or progesterone (PG) may have cardioprotective effects, recent controversy regarding hormone replacement therapy has questioned the cardiac effects of these steroids. Using cardiomyocytes, we tested whether ES or PG has protective effects at the cellular level. We found that PG but not ES protects cardiomyocytes from apoptotic cell death induced by doxorubicin (Dox). PG inhibited apoptosis in a dose-dependent manner, by 12 ± 4.0% at 1 μm and 60 ± 1.0% at 10 μm. The anti-apoptotic effect of PG was also time dependent, causing 18 ± 5% or 62 + 2% decrease in caspase-3 activity within 1 h or 72 h of pretreatment. While PG causes nuclear translocation of its receptor within 20 min, the cytoprotective effect of PG was canceled by mifepristone (MF), a PG receptor antagonist. Analyses using Affymetrix high-density oligonucleotide array and RT-PCR found that PG induced Bcl-xL, metallothionine, NADPH quinone oxidoreductase 1, glutathione peroxidase-3, and four isoforms of glutathione S-transferase. Western blot analyses revealed that PG indeed induced an elevation of Bcl-xL protein in a dose- and time-dependent manner. Nuclear run-on assay indicated that PG induced Bcl-xL gene transcription. Inhibiting the expression of Bcl-xL using siRNA reduced the cytoprotective effect of PG. Our data suggests that PG induces a cytoprotective effect in cardiomyocytes in association with induction of Bcl-xL gene.
Collapse
Affiliation(s)
- Stephen Morrissy
- Department of Pharmacology, University of Arizona, Tucson, 85724, USA
| | | | | | | | | |
Collapse
|
198
|
Sildenafil increases chemotherapeutic efficacy of doxorubicin in prostate cancer and ameliorates cardiac dysfunction. Proc Natl Acad Sci U S A 2010; 107:18202-7. [PMID: 20884855 DOI: 10.1073/pnas.1006965107] [Citation(s) in RCA: 127] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
We have shown that the potent phosphodiesterase-5 (PDE-5) inhibitor sildenafil (Viagra) induces a powerful effect on reduction of infarct size following ischemia/reperfusion injury and improvement of left ventricular dysfunction in the failing heart after myocardial infarction or doxorubicin (DOX) treatment. In the present study, we further investigated the potential effects of sildenafil on improving antitumor efficacy of DOX in prostate cancer. Cotreatment with sildenafil enhanced DOX-induced apoptosis in PC-3 and DU145 prostate cancer cells, which was mediated by enhanced generation of reactive oxygen species, up-regulation of caspase-3 and caspase-9 activities, reduced expression of Bcl-xL, and phosphorylation of Bad. Overexpression of Bcl-xL or dominant negative caspase 9 attenuated the synergistic effect of sildenafil and DOX on prostate cancer cell killing. Furthermore, treatment with sildenafil and DOX in mice bearing prostate tumor xenografts resulted in significant inhibition of tumor growth. The reduced tumor size was associated with amplified apoptotic cell death and increased expression of activated caspase 3. Doppler echocardiography showed that sildenafil treatment ameliorated DOX-induced left ventricular dysfunction. In conclusion, these results provide provocative evidence that sildenafil is both a powerful sensitizer of DOX-induced killing of prostate cancer while providing concurrent cardioprotective benefit.
Collapse
|
199
|
Cardiac toxicity in breast cancer patients: from a fractional point of view to a global assessment. Cancer Treat Rev 2010; 37:321-30. [PMID: 20864260 DOI: 10.1016/j.ctrv.2010.08.007] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2010] [Revised: 08/19/2010] [Accepted: 08/20/2010] [Indexed: 11/20/2022]
Abstract
When focusing on heart disease, most available studies split the two different parts of the adjuvant treatment, i.e., systemic therapies and radiation therapy, making it difficult to implement efficient strategies for preventing treatment-induced cardiac toxicity. This paper reviews the current understanding of treatments-induced cardiac toxicity in a global approach. Many factors should be considered when assessing the cardiac hazard. Treatment-related risk factors include heart dose exposure, chemotherapy, targeted agents such as HER2 inhibitors, but also endocrine agents, or anesthetic procedure. Patients' characteristics should also be taken into account. Age, menopausal status, stress, previous history of cardiac disease, genetic profile, and body mass index could all impact on cardiac function after adjuvant therapies. Cardiac toxicity should not be analyzed as the consequence of a specific therapy, but should be considered as the result of additive or supra-additive toxicities. By this way, it will be possible to implement new strategies for preventing treatment-induced cardiac toxicity.
Collapse
|
200
|
Kavazis AN, Smuder AJ, Min K, Tümer N, Powers SK. Short-term exercise training protects against doxorubicin-induced cardiac mitochondrial damage independent of HSP72. Am J Physiol Heart Circ Physiol 2010; 299:H1515-24. [PMID: 20833957 DOI: 10.1152/ajpheart.00585.2010] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Doxorubicin (Dox) is an antitumor agent used in cancer treatment, but its clinical use is limited due to cardiotoxicity. Although exercise training can defend against Dox-mediated cardiac damage, the means for this cardioprotection remain unknown. To investigate the mechanism(s) responsible for exercise training-induced cardioprotection against Dox-mediated cardiotoxicity, we tested a two-pronged hypothesis: 1) exercise training protects against Dox-induced cardiotoxicity by preventing Dox-mediated mitochondrial damage/dysfunction and increased oxidative stress and 2) exercise training-induced cardiac expression of the inducible isoform of the 70-kDa heat shock protein 72 (HSP72) is essential to achieve exercise training-induced cardioprotection against Dox toxicity. Animals were randomly assigned to sedentary or exercise groups and paired with either placebo or Dox treatment (i.e., 20 mg/kg body wt ip Dox hydrochloride 24 h before euthanasia). Dox administration resulted in cardiac mitochondrial dysfunction, activation of proteases, and apoptosis. Exercise training increased cardiac antioxidant enzymes and HSP72 protein abundance and protected cardiac myocytes against Dox-induced mitochondrial damage, protease activation, and apoptosis. To determine whether exercise-induced expression of HSP72 in the heart is required for this cardioprotection, we utilized an innovative experimental strategy that successfully prevented exercise-induced increases in myocardial HSP72 levels. However, prevention of exercise-induced increases in myocardial HSP72 did not eliminate the exercise-induced cardioprotective phenotype that is resistant to Dox-mediated injury. Our results indicate that exercise training protects against the detrimental side effects of Dox in cardiac myocytes, in part, by protecting mitochondria against Dox-mediated damage. However, this exercise-induced cardioprotection is independent of myocardial HSP72 levels. Finally, our data are consistent with the concept that increases in cardiac mitochondrial antioxidant enzymes may contribute to exercise-induced cardioprotection.
Collapse
|