1
|
Ali MA, Zhang Y, Ni Q, Huang AQ, Dou S, Tang Y, Xu X, Zeng C. SIRT5 alleviates oxidative stress of boar sperm induced by cryopreservation through IDH2 and SOD2 pathway. Theriogenology 2025; 241:117424. [PMID: 40198939 DOI: 10.1016/j.theriogenology.2025.117424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 04/02/2025] [Accepted: 04/03/2025] [Indexed: 04/10/2025]
Abstract
Excessive production of reactive oxygen species (ROS) and lipid peroxidation result in decreased motility, fertility, farrowing rate and litter size during boar sperm cryopreservation. Our previous study reported that cryopreservation leads to differential expression of genes (SIRT5 and IDH2) which regulate the energy metabolism and quality of sperm. But how SIRT5 regulates the metabolism and oxidative stress in boar sperm are still unknown. In the present study, we first determined the regulation of IDH2 and SOD2 by SIRT5 after its knockdown and then determined the role of SIRT5 in regulation of oxidative stress, metabolic activities, motility and fertility potential by its specific inhibitor (Suramin, SUR) and activator (Resveratrol, RSV). Then the effect of cryopreservation was evaluated on mRNA and protein expressions of SIRT5, IDH2 and SOD2 in boar sperm. A decrease in expressions was observed at both mRNA and protein for all these genes. Additionally, RSV and SUR treatment led to up and downregulation of mRNA and protein of both IDH2 and SOD2. A significant decrease (P < 0.05) in ROS levels and improvement in sperm motility, acrosomal integrity, MMP, PMI and ATP were observed in post-thaw sperm after activation of SIRT5 with RSV. Additionally, BAX and 4HNE levels were significantly (P < 0.05) reduced in RSV treated sperm. In conclusion, this study demonstrated that SIRT5 activator (RSV) improved the quality of boar sperm by upregulation of IDH2 and SOD2. At the same time, the oxidative stress was reduced via interaction among SIRT5, IDH2 and SOD2.
Collapse
Affiliation(s)
- Malik Ahsan Ali
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China; Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China; College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China; Key Laboratory of Animal Genetics, Breeding and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Yan Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China; Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China; College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Qingyong Ni
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China; Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China; College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - An-Qi Huang
- College of Life Science, Sichuan Agricultural University, Ya'an, 625014, China
| | - Shan Dou
- College of Life Science, Sichuan Agricultural University, Ya'an, 625014, China
| | - Yiguo Tang
- Daocheng Kangbaqing Animal Husbandry Co., Ltd, Sichuan, China
| | - Xinhong Xu
- Daocheng Kangbaqing Animal Husbandry Co., Ltd, Sichuan, China
| | - Changjun Zeng
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China; Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China; College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China.
| |
Collapse
|
2
|
Gao H, Chen Z, Yao Y, He Y, Hu X. Common biological processes and mutual crosstalk mechanisms between cardiovascular disease and cancer. Front Oncol 2024; 14:1453090. [PMID: 39634266 PMCID: PMC11614734 DOI: 10.3389/fonc.2024.1453090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 10/31/2024] [Indexed: 12/07/2024] Open
Abstract
Cancer and cardiovascular disease (CVD) are leading causes of mortality and thus represent major health challenges worldwide. Clinical data suggest that cancer patients have an increased likelihood of developing cardiovascular disease, while epidemiologic studies have shown that patients with cardiovascular disease are also more likely to develop cancer. These observations underscore the increasing importance of studies exploring the mechanisms underlying the interaction between the two diseases. We review their common physiological processes and potential pathophysiological links. We explore the effects of chronic inflammation, oxidative stress, and disorders of fatty acid metabolism in CVD and cancer, and also provide insights into how cancer and its treatments affect heart health, as well as present recent advances in reverse cardio-oncology using a new classification approach.
Collapse
Affiliation(s)
- Hanwei Gao
- Department of Cardiology, China–Japan Union Hospital of Jilin University, Jilin University, Changchun, Jilin, China
| | - Zhongyu Chen
- Department of Cardiology, China–Japan Union Hospital of Jilin University, Jilin University, Changchun, Jilin, China
- CJUH-JLU-China iGEM Team, Jilin University, Changchun, Jilin, China
| | - Yutong Yao
- Department of Cardiology, China–Japan Union Hospital of Jilin University, Jilin University, Changchun, Jilin, China
- CJUH-JLU-China iGEM Team, Jilin University, Changchun, Jilin, China
| | - Yuquan He
- Department of Cardiology, China–Japan Union Hospital of Jilin University, Jilin University, Changchun, Jilin, China
- CJUH-JLU-China iGEM Team, Jilin University, Changchun, Jilin, China
| | - Xin Hu
- Department of Cardiology, China–Japan Union Hospital of Jilin University, Jilin University, Changchun, Jilin, China
- CJUH-JLU-China iGEM Team, Jilin University, Changchun, Jilin, China
| |
Collapse
|
3
|
Zhao P, Guo C, Du H, Xiao Y, Su J, Wang X, Yeung WSB, Li G, Wang T. Chemotherapy-induced ovarian damage and protective strategies. HUM FERTIL 2023; 26:887-900. [PMID: 38054300 DOI: 10.1080/14647273.2023.2275764] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 10/14/2023] [Indexed: 12/07/2023]
Abstract
More than 9.2 million women worldwide suffer from cancer, and about 5% of them are at reproductive age. Chemotherapy-induced impairment of fertility affects the quality of life of these women. Several chemotherapeutic agents have been proven to cause apoptosis and autophagy by inducing DNA damage and cellular stress. Injuries to the ovarian stroma and micro-vessel network are also considered as pivotal factors resulting in ovarian dysfunction induced by chemotherapeutic agents. Primordial follicle pool over-activation may also be the mechanism inducing damage to the ovarian reserve. Although many studies have explored the mechanisms involved in chemotherapy-induced reproductive toxicity, the exact molecular mechanisms have not been elucidated. It is essential to understand the mechanisms involved in ovarian damage, in order to develop potential protective treatments to preserve fertility. In this article, we reviewed the current knowledge on the mechanism of chemotherapy-induced ovarian damage and possible protective strategies that prevent the ovary from such damages.
Collapse
Affiliation(s)
- Peikun Zhao
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, PR China
| | - Chenxi Guo
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, PR China
| | - Huijia Du
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, PR China
| | - Yuan Xiao
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, PR China
| | - Jiaping Su
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, PR China
| | - Xiaohui Wang
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, PR China
| | - Willian S B Yeung
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, PR China
| | - Guangxin Li
- Department of Breast and Thyroid Surgery, Peking University Shenzhen Hospital, Shenzhen, PR China
| | - Tianren Wang
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, PR China
| |
Collapse
|
4
|
Extracellular Vesicles and Cancer Therapy: Insights into the Role of Oxidative Stress. Antioxidants (Basel) 2022; 11:antiox11061194. [PMID: 35740091 PMCID: PMC9228181 DOI: 10.3390/antiox11061194] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/13/2022] [Accepted: 06/15/2022] [Indexed: 02/04/2023] Open
Abstract
Oxidative stress plays a significant role in cancer development and cancer therapy, and is a major contributor to normal tissue injury. The unique characteristics of extracellular vesicles (EVs) have made them potentially useful as a diagnostic tool in that their molecular content indicates their cell of origin and their lipid membrane protects the content from enzymatic degradation. In addition to their possible use as a diagnostic tool, their role in how normal and diseased cells communicate is of high research interest. The most exciting area is the association of EVs, oxidative stress, and pathogenesis of numerous diseases. However, the relationship between oxidative stress and oxidative modifications of EVs is still unclear, which limits full understanding of the clinical potential of EVs. Here, we discuss how EVs, oxidative stress, and cancer therapy relate to one another; how oxidative stress can contribute to the generation of EVs; and how EVs’ contents reveal the presence of oxidative stress. We also point out the potential promise and limitations of using oxidatively modified EVs as biomarkers of cancer and tissue injury with a focus on pediatric oncology patients.
Collapse
|
5
|
Chaiswing L, Xu F, Zhao Y, Thorson J, Wang C, He D, Lu J, Ellingson SR, Zhong W, Meyer K, Luo W, St. Clair W, Clair DS. The RelB-BLNK Axis Determines Cellular Response to a Novel Redox-Active Agent Betamethasone during Radiation Therapy in Prostate Cancer. Int J Mol Sci 2022; 23:ijms23126409. [PMID: 35742868 PMCID: PMC9223669 DOI: 10.3390/ijms23126409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/01/2022] [Accepted: 06/04/2022] [Indexed: 11/16/2022] Open
Abstract
Aberrant levels of reactive oxygen species (ROS) are potential mechanisms that contribute to both cancer therapy efficacy and the side effects of cancer treatment. Upregulation of the non-canonical redox-sensitive NF-kB family member, RelB, confers radioresistance in prostate cancer (PCa). We screened FDA-approved compounds and identified betamethasone (BET) as a drug that increases hydrogen peroxide levels in vitro and protects non-PCa tissues/cells while also enhancing radiation killing of PCa tissues/cells, both in vitro and in vivo. Significantly, BET increases ROS levels and exerts different effects on RelB expression in normal cells and PCa cells. BET induces protein expression of RelB and RelB target genes, including the primary antioxidant enzyme, manganese superoxide dismutase (MnSOD), in normal cells, while it suppresses protein expression of RelB and MnSOD in LNCaP cells and PC3 cells. RNA sequencing analysis identifies B-cell linker protein (BLNK) as a novel RelB complementary partner that BET differentially regulates in normal cells and PCa cells. RelB and BLNK are upregulated and correlate with the aggressiveness of PCa in human samples. The RelB-BLNK axis translocates to the nuclear compartment to activate MnSOD protein expression. BET promotes the RelB-BLNK axis in normal cells but suppresses the RelB-BLNK axis in PCa cells. Targeted disruptions of RelB-BLNK expressions mitigate the radioprotective effect of BET on normal cells and the radiosensitizing effect of BET on PCa cells. Our study identified a novel RelB complementary partner and reveals a complex redox-mediated mechanism showing that the RelB-BLNK axis, at least in part, triggers differential responses to the redox-active agent BET by stimulating adaptive responses in normal cells but pushing PCa cells into oxidative stress overload.
Collapse
Affiliation(s)
- Luksana Chaiswing
- Department of Toxicology and Cancer Biology, University of Kentucky, 452 Health Sciences Research Building, Lexington, KY 40536, USA; (F.X.); (Y.Z.)
- Correspondence: (L.C.); (D.S.C.)
| | - Fangfang Xu
- Department of Toxicology and Cancer Biology, University of Kentucky, 452 Health Sciences Research Building, Lexington, KY 40536, USA; (F.X.); (Y.Z.)
| | - Yanming Zhao
- Department of Toxicology and Cancer Biology, University of Kentucky, 452 Health Sciences Research Building, Lexington, KY 40536, USA; (F.X.); (Y.Z.)
| | - Jon Thorson
- Center for Pharmaceutical Research and Innovation, Lexington, KY 40536, USA;
- College of Pharmacy, Pharmaceutical Sciences Department, University of Kentucky, Lexington, KY 40536, USA
| | - Chi Wang
- Markey Biostatistics and Bioinformatics Shared Resource Facility, University of Kentucky, Lexington, KY 40536, USA; (C.W.); (D.H.); (J.L.); (S.R.E.)
| | - Daheng He
- Markey Biostatistics and Bioinformatics Shared Resource Facility, University of Kentucky, Lexington, KY 40536, USA; (C.W.); (D.H.); (J.L.); (S.R.E.)
| | - Jinpeng Lu
- Markey Biostatistics and Bioinformatics Shared Resource Facility, University of Kentucky, Lexington, KY 40536, USA; (C.W.); (D.H.); (J.L.); (S.R.E.)
| | - Sally R. Ellingson
- Markey Biostatistics and Bioinformatics Shared Resource Facility, University of Kentucky, Lexington, KY 40536, USA; (C.W.); (D.H.); (J.L.); (S.R.E.)
| | - Weixiong Zhong
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI 53705, USA; (W.Z.); (K.M.)
| | - Kristy Meyer
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI 53705, USA; (W.Z.); (K.M.)
| | - Wei Luo
- Department of Radiation Medicine, University of Kentucky, Lexington, KY 40536, USA; (W.L.); (W.S.C.)
| | - William St. Clair
- Department of Radiation Medicine, University of Kentucky, Lexington, KY 40536, USA; (W.L.); (W.S.C.)
| | - Daret St. Clair
- Department of Toxicology and Cancer Biology, University of Kentucky, 452 Health Sciences Research Building, Lexington, KY 40536, USA; (F.X.); (Y.Z.)
- Correspondence: (L.C.); (D.S.C.)
| |
Collapse
|
6
|
Liao W, Rao Z, Wu L, Chen Y, Li C. Cariporide Attenuates Doxorubicin-Induced Cardiotoxicity in Rats by Inhibiting Oxidative Stress, Inflammation and Apoptosis Partly Through Regulation of Akt/GSK-3β and Sirt1 Signaling Pathway. Front Pharmacol 2022; 13:850053. [PMID: 35747748 PMCID: PMC9209753 DOI: 10.3389/fphar.2022.850053] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 04/12/2022] [Indexed: 11/24/2022] Open
Abstract
Background: Doxorubicin (DOX) is a potent chemotherapeutic agent with limited usage due to its cumulative cardiotoxicity. The Na+/H+ exchanger isoform 1 (NHE1) is a known regulator of oxidative stress, inflammation, and apoptosis. The present study was designed to investigate the possible protective effect of cariporide (CAR), a selective inhibitor of NHE1, against DOX-induced cardiotoxicity in rats. Methods: Male Sprague-Dawley rats were intraperitoneally injected with DOX to induce cardiac toxicity and CAR was given orally for treatment. The injured H9c2 cell model was established by incubation with DOX in vitro. Echocardiography, as well as morphological and ultra-structural examination were performed to evaluate cardiac function and histopathological changes. The biochemical parameters were determined according to the manufacturer’s guideline of kits. ROS were assessed by using an immunofluorescence assay. The serum levels and mRNA expressions of inflammatory cytokines were measured by using ELISA or qRT-PCR. Cardiac cell apoptosis and H9c2 cell viability were tested by TUNEL or MTT method respectively. The protein expressions of Cleaved-Caspase-3, Bcl-2, Bax, Akt, GSK-3β, and Sirt1 were detected by western blot. Results: Treatment with CAR protected against DOX-induced body weight changes, impairment of heart function, leakage of cardiac enzymes, and heart histopathological damage. In addition, CAR significantly attenuated oxidative stress and inhibited the levels and mRNA expressions of inflammatory cytokines (TNF-α, IL-6, IL-18, and IL-1β), which were increased by DOX treatment. Moreover, CAR significantly suppressed myocardial apoptosis and Cleaved-Caspase-3 protein expression induced by DOX, which was in agreement with the increased Bcl-2/Bax ratio. Also, DOX suppressed phosphorylation of Akt and GSK-3β, which was significantly reversed by administration of CAR. Furthermore, CAR treatment prevented DOX-induced down-regulation of Sirt1 at the protein level in vitro and in vivo. Finally, Sirt1 inhibitor reversed the protective effects of CAR, as evidenced by reduced cell viability and Sirt1 protein expression in vitro. Conclusion: Taken together, we provide evidence for the first time in the current study that CAR exerts potent protective effects against DOX-induced cardiotoxicity in rats. This cardio-protective effect is attributed to suppressing oxidative stress, inflammation, and apoptosis, at least in part, through regulation of Akt/GSK-3β and Sirt1 signaling pathway, which has not been reported to date.
Collapse
Affiliation(s)
- Wenli Liao
- National Demonstration Center for Experimental General Medicine Education, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Zhiwei Rao
- Central Hospital of Xianning, The First Affiliate Hospital of Hubei University of Science and Technology, Xianning, China
| | - Lingling Wu
- National Demonstration Center for Experimental General Medicine Education, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Yong Chen
- Central Hospital of Xianning, The First Affiliate Hospital of Hubei University of Science and Technology, Xianning, China
- *Correspondence: Cairong Li, ; Yong Chen,
| | - Cairong Li
- National Demonstration Center for Experimental General Medicine Education, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
- *Correspondence: Cairong Li, ; Yong Chen,
| |
Collapse
|
7
|
Yang HB, Lu ZY, Yuan W, Li WD, Mao S. Selenium Attenuates Doxorubicin-Induced Cardiotoxicity Through Nrf2-NLRP3 Pathway. Biol Trace Elem Res 2022; 200:2848-2856. [PMID: 34462843 DOI: 10.1007/s12011-021-02891-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 08/13/2021] [Indexed: 12/19/2022]
Abstract
Selenium (Se), an essential nutrient for humans, has been reported to possess cardioprotective effect. However, the protective effects of Se against doxorubicin (DOX)-induced cardiotoxicity and the underlying mechanism are rarely reported. In this study, we sought to explore whether Se protected against DOX-induced cardiotoxicity by inhibiting Nrf2-NLRP3 pathway. We found that Se treatment effectively alleviated DOX-induced myocardial dysfunctions, decreasing plasma markers associated with myocardial injury. Moreover, Se treatment significantly inhibited DOX-induced oxidative damages and pro-inflammatory cytokine expression in heart tissues. Furthermore, Se treatment markedly promoted the expression of Nrf2 and prevented the activation of NLRP3 inflammasome. Importantly, suppression of Nrf2 abolished the cardioprotective effects of Se and diminished the inhibition of Se on NLRP3 inflammasome. Collectively, our study demonstrated that Se might protect against DOX-induced cardiotoxicity via regulating Nrf2-NLRP3 pathway. Se supplementation may be a potential therapeutic strategy to protect against DOX-induced cardiac injury.
Collapse
Affiliation(s)
- Hai-Bing Yang
- Department of Cardiology, Yingshang First Hospital, Yingli Road, Fuyang, 236000, China.
| | - Zhao-Yang Lu
- Department of Cardiology, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, China.
| | - Wei Yuan
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Jie Fang Road 438, Zhenjiang, 212001, China
| | - Wei-Dong Li
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Jie Fang Road 438, Zhenjiang, 212001, China
| | - Shang Mao
- Department of Cardiology, Yingshang First Hospital, Yingli Road, Fuyang, 236000, China
| |
Collapse
|
8
|
Kitakata H, Endo J, Ikura H, Moriyama H, Shirakawa K, Katsumata Y, Sano M. Therapeutic Targets for DOX-Induced Cardiomyopathy: Role of Apoptosis vs. Ferroptosis. Int J Mol Sci 2022; 23:1414. [PMID: 35163335 PMCID: PMC8835899 DOI: 10.3390/ijms23031414] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/20/2022] [Accepted: 01/24/2022] [Indexed: 01/04/2023] Open
Abstract
Doxorubicin (DOX) is the most widely used anthracycline anticancer agent; however, its cardiotoxicity limits its clinical efficacy. Numerous studies have elucidated the mechanisms underlying DOX-induced cardiotoxicity, wherein apoptosis has been reported as the most common final step leading to cardiomyocyte death. However, in the past two years, the involvement of ferroptosis, a novel programmed cell death, has been proposed. The purpose of this review is to summarize the historical background that led to each form of cell death, focusing on DOX-induced cardiotoxicity and the molecular mechanisms that trigger each form of cell death. Furthermore, based on this understanding, possible therapeutic strategies to prevent DOX cardiotoxicity are outlined. DNA damage, oxidative stress, intracellular signaling, transcription factors, epigenetic regulators, autophagy, and metabolic inflammation are important factors in the molecular mechanisms of DOX-induced cardiomyocyte apoptosis. Conversely, the accumulation of lipid peroxides, iron ion accumulation, and decreased expression of glutathione and glutathione peroxidase 4 are important in ferroptosis. In both cascades, the mitochondria are an important site of DOX cardiotoxicity. The last part of this review focuses on the significance of the disruption of mitochondrial homeostasis in DOX cardiotoxicity.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Motoaki Sano
- Department of Cardiology, Keio University School of Medicine, Tokyo 160-8582, Japan; (H.K.); (J.E.); (H.I.); (H.M.); (K.S.); (Y.K.)
| |
Collapse
|
9
|
Chen Y, Wang L, Liu T, Qiu Z, Qiu Y, Liu D. Inhibitory effects of Panax ginseng glycoproteins in models of doxorubicin-induced cardiac toxicity in vivo and in vitro. Food Funct 2021; 12:10862-10874. [PMID: 34617939 DOI: 10.1039/d1fo01307f] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Doxorubicin (DOX) is an effective antineoplastic drug; however, its clinical application is limited owing to the side effect of fatal heart dysfunction on its use. Panax ginseng glycoproteins have antioxidant, antiapoptotic, and anti-inflammatory properties. Thus, the aim of this study was to investigate the effects and possible action mechanisms of P. ginseng glycoproteins against DOX-induced cardiotoxicity. To this end, we used an in vitro model of DOX-treated H9C2 cells and an in vivo model of DOX-treated rats. We found that P. ginseng glycoproteins markedly increased H9C2 cell viability, decreased creatine kinase and lactate dehydrogenase levels, and improved histopathological and electrocardiogram changes in rats, protecting them from DOX-induced cardiotoxicity. Furthermore, P. ginseng glycoproteins significantly inhibited myocardial oxidative insult through adjusting the intracellular ROS, MDA, SOD, and GSH levels in vitro and in vivo. In conclusion, our data suggest that P. ginseng glycoproteins alleviated DOX-induced myocardial oxidative stress-related cardiotoxicity. This natural product could be developed as a new candidate for alleviating DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Yajun Chen
- Department of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China.
| | - Lei Wang
- Department of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China.
| | - Tianjia Liu
- Department of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China.
| | - Zhidong Qiu
- Department of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China.
| | - Ye Qiu
- Department of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China.
| | - Da Liu
- Department of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China.
| |
Collapse
|
10
|
Koss-Mikołajczyk I, Todorovic V, Sobajic S, Mahajna J, Gerić M, Tur JA, Bartoszek A. Natural Products Counteracting Cardiotoxicity during Cancer Chemotherapy: The Special Case of Doxorubicin, a Comprehensive Review. Int J Mol Sci 2021; 22:10037. [PMID: 34576204 PMCID: PMC8467966 DOI: 10.3390/ijms221810037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/14/2021] [Accepted: 09/15/2021] [Indexed: 12/14/2022] Open
Abstract
Cardiotoxicity is a frequent undesirable phenomenon observed during oncological treatment that limits the therapeutic dose of antitumor drugs and thus may decrease the effectiveness of cancer eradication. Almost all antitumor drugs exhibit toxic properties towards cardiac muscle. One of the underlying causes of cardiotoxicity is the stimulation of oxidative stress by chemotherapy. This suggests that an appropriately designed diet or dietary supplements based on edible plants rich in antioxidants could decrease the toxicity of antitumor drugs and diminish the risk of cardiac failure. This comprehensive review compares the cardioprotective efficacy of edible plant extracts and foodborne phytochemicals whose beneficial activity was demonstrated in various models in vivo and in vitro. The studies selected for this review concentrated on a therapy frequently applied in cancer, anthracycline antibiotic-doxorubicin-as the oxidative stress- and cardiotoxicity-inducing agent.
Collapse
Affiliation(s)
- Izabela Koss-Mikołajczyk
- Department of Food Chemistry, Technology and Biotechnology, Gdańsk University of Technology, 11/12 Narutowicza St., 80-233 Gdańsk, Poland;
| | - Vanja Todorovic
- Department of Bromatology, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia; (V.T.); (S.S.)
| | - Sladjana Sobajic
- Department of Bromatology, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia; (V.T.); (S.S.)
| | - Jamal Mahajna
- Department of Nutrition and Natural Products, Migal-Galilee Research Institute, Kiryat Shmona 11016, Israel;
- Department of Nutritional Sciences, Tel-Hai College, Qiryat Shemona 1220800, Israel
| | - Marko Gerić
- Mutagenesis Unit, Institute for Medical Research and Occupational Health, 10000 Zagreb, Croatia;
| | - Josep A. Tur
- Research Group on Community Nutrition & Oxidative Stress, University of Balearic Islands—IUNICS, IDISBA & CIBEROBN (Physiopathology of Obesity and Nutrition), 07122 Palma de Mallorca, Spain;
| | - Agnieszka Bartoszek
- Department of Food Chemistry, Technology and Biotechnology, Gdańsk University of Technology, 11/12 Narutowicza St., 80-233 Gdańsk, Poland;
| |
Collapse
|
11
|
AQP3 and AQP5-Potential Regulators of Redox Status in Breast Cancer. Molecules 2021; 26:molecules26092613. [PMID: 33947079 PMCID: PMC8124745 DOI: 10.3390/molecules26092613] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/22/2021] [Accepted: 04/27/2021] [Indexed: 12/23/2022] Open
Abstract
Breast cancer is still one of the leading causes of mortality in the female population. Despite the campaigns for early detection, the improvement in procedures and treatment, drastic improvement in survival rate is omitted. Discovery of aquaporins, at first described as cellular plumbing system, opened new insights in processes which contribute to cancer cell motility and proliferation. As we discover new pathways activated by aquaporins, the more we realize the complexity of biological processes and the necessity to fully understand the pathways affected by specific aquaporin in order to gain the desired outcome-remission of the disease. Among the 13 human aquaporins, AQP3 and AQP5 were shown to be significantly upregulated in breast cancer indicating their role in the development of this malignancy. Therefore, these two aquaporins will be discussed for their involvement in breast cancer development, regulation of oxidative stress and redox signalling pathways leading to possibly targeting them for new therapies.
Collapse
|
12
|
Ansheles AA, Sergienko IV, Prus YA, Sergienko VB. Nuclear imaging of chemotherapy-induced cardiotoxicity. КАРДИОВАСКУЛЯРНАЯ ТЕРАПИЯ И ПРОФИЛАКТИКА 2021. [DOI: 10.15829/1728-8800-2021-2537] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The high efficiency of modern chemotherapy has made it possible to achieve great success in the treatment of cancer. Cardiovascular adverse effects are a major disadvantage of anticancer therapy, often requiring low and less effective doses or even drug withdrawal. Nuclear imaging techniques are the most sensitive in early detection of left ventricular damage and dysfunction during chemotherapy. This review presents modern data on the potential of nuclear imaging of cardiotoxicity.
Collapse
Affiliation(s)
| | | | - Yu. A. Prus
- National Medical Research Center of Cardiology
| | | |
Collapse
|
13
|
Alzahrani AM, Rajendran P, Veeraraghavan VP, Hanieh H. Cardiac Protective Effect of Kirenol against Doxorubicin-Induced Cardiac Hypertrophy in H9c2 Cells through Nrf2 Signaling via PI3K/AKT Pathways. Int J Mol Sci 2021; 22:ijms22063269. [PMID: 33806909 PMCID: PMC8004766 DOI: 10.3390/ijms22063269] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/08/2021] [Accepted: 03/09/2021] [Indexed: 12/14/2022] Open
Abstract
Kirenol (KRL) is a biologically active substance extracted from Herba Siegesbeckiae. This natural type of diterpenoid has been widely adopted for its important anti-inflammatory and anti-rheumatic properties. Despite several studies claiming the benefits of KRL, its cardiac effects have not yet been clarified. Cardiotoxicity remains a key concern associated with the long-term administration of doxorubicin (DOX). The generation of reactive oxygen species (ROS) causes oxidative stress, significantly contributing to DOX-induced cardiac damage. The purpose of the current study is to investigate the cardio-protective effects of KRL against apoptosis in H9c2 cells induced by DOX. The analysis of cellular apoptosis was performed using the terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) staining assay and measuring the modulation in the expression levels of proteins involved in apoptosis and Nrf2 signaling, the oxidative stress markers. Furthermore, Western blotting was used to determine cell survival. KRL treatment, with Nrf2 upregulation and activation, accompanied by activation of PI3K/AKT, could prevent the administration of DOX to induce cardiac oxidative stress, remodeling, and other effects. Additionally, the diterpenoid enhanced the activation of Bcl2 and Bcl-xL, while suppressing apoptosis marker proteins. As a result, KRL is considered a potential agent against hypertrophy resulting from cardiac deterioration. The study results show that KRL not only activates the IGF-IR-dependent p-PI3K/p-AKT and Nrf2 signaling pathway, but also suppresses caspase-dependent apoptosis.
Collapse
Affiliation(s)
- Abdullah M. Alzahrani
- Department of Biological Sciences, College of Science, King Faisal University, Al Ahsa 31982, Saudi Arabia;
| | - Peramaiyan Rajendran
- Department of Biological Sciences, College of Science, King Faisal University, Al Ahsa 31982, Saudi Arabia;
- Correspondence: ; Tel.: +97-0135899543
| | - Vishnu Priya Veeraraghavan
- Department of Biochemistry, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600 077, India;
| | - Hamza Hanieh
- Department of Medical Analysis, Al-Hussein Bin Talal University, Ma’an 71111, Jordan;
| |
Collapse
|
14
|
Activation of Nrf2 by miR-152 Inhibits Doxorubicin-Induced Cardiotoxicity via Attenuation of Oxidative Stress, Inflammation, and Apoptosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8860883. [PMID: 33574984 PMCID: PMC7857911 DOI: 10.1155/2021/8860883] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 12/23/2020] [Accepted: 01/11/2021] [Indexed: 12/18/2022]
Abstract
Doxorubicin (DOX) could trigger congestive heart failure, which largely limited the clinical use of DOX. microRNAs (miRNAs) were closely involved in the pathogenesis of DOX-induced cardiomyopathy. Here, we aimed to investigate the effect of miR-152 on DOX-induced cardiotoxicity in mice. To study this, we used an adeno-associated viral vector to overexpress miR-152 in mice 6 weeks before DOX treatment, using a dose mimicking the concentrations used in the clinics. In response to DOX injection, miR-152 was significantly decreased in murine hearts and cardiomyocytes. After DOX treatment, mice with miR-152 overexpression in the hearts developed less cardiac dysfunction, oxidative stress, inflammation, and myocardial apoptosis. Furthermore, we found that miR-152 overexpression attenuated DOX-related oxidative stress, inflammation, and cell loss in cardiomyocytes, whereas miR-152 knockdown resulted in oxidative stress, inflammation, and cell loss in cardiomyocytes. Mechanistically, this effect of miR-152 was dependent on the activation of nuclear factor (erythroid-derived 2)-like 2 (Nrf2) in response to DOX. Notably, Nrf2 deficiency blocked the protective effects of miR-152 against DOX-related cardiac injury in mice. In conclusion, miR-152 protected against DOX-induced cardiotoxicity via the activation of the Nrf2 signaling pathway. These results suggest that miR-152 may be a promising therapeutic target for the treatment of DOX-induced cardiotoxicity.
Collapse
|
15
|
Human Amnion Membrane Proteins Prevent Doxorubicin-Induced Oxidative Stress Injury and Apoptosis in Rat H9c2 Cardiomyocytes. Cardiovasc Toxicol 2021; 20:370-379. [PMID: 32086724 DOI: 10.1007/s12012-020-09564-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Doxorubicin (DOX) is widely used as an effective chemotherapy agent in cancer treatment. Cardiac toxicity in cancer treatment with DOX demand urgent attention and no effective treatment has been established for DOX-induced cardiomyopathy. It has been well documented that human amniotic membrane proteins (AMPs), extracted from amnion membrane (AM), have antioxidant, anti-apoptotic, and cytoprotective properties. Therefore, in this study, we aimed to investigate the protective effects of AMPs against cardiotoxicity induced by DOX in cultured rat cardiomyocyte cells (H9c2). DOX-induced cell injury was evaluated using multi-parametric assay including thiazolyl blue tetrazolium bromide (MTT), the release of lactic dehydrogenase (LDH), intracellular Ca2+ , reactive oxygen species (ROS) levels, cellular antioxidant status, mitochondrial membrane potential (ΔΨm), malondialdehyde (MDA), and NF-κB p65 DNA-binding activity. Moreover, expression profiling of apoptosis-related genes (P53, Bcl-2, and Bax) and Annexin V by flow cytometry were used for cell apoptosis detection. It was shown that AMPs pretreatment inhibited the cell toxicity induced by DOX. AMPs effectively attenuated the increased levels of LDH, Ca2+ , ROS, and MDA and also simultaneously elevated the ΔΨm and antioxidant status such as superoxide dismutase (SOD) and Catalase (CAT) in pretreated H9c2 cardiomyocytes. Besides, the activity of NF-kB p65 was reduced and the p53 and Bax protein levels were inhibited in these myocardial cells subjected to DOX. These findings provide the first evidence that AMPs potently suppressed DOX-induced toxicity in cardiomyocytes through inhibition of oxidative stress and apoptosis. Thus, AMPs can be a potential therapeutic agent against DOX cardiotoxicity.
Collapse
|
16
|
Cheong A, McGrath S, Robinson T, Maliki R, Spurling A, Lock P, Rephaeli A, Nudelman A, Parker BS, Pepe S, Cutts SM. A switch in mechanism of action prevents doxorubicin-mediated cardiac damage. Biochem Pharmacol 2021; 185:114410. [PMID: 33428897 DOI: 10.1016/j.bcp.2021.114410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/03/2021] [Accepted: 01/05/2021] [Indexed: 10/22/2022]
Abstract
Cancer patients treated with doxorubicin are at risk of congestive heart failure due to doxorubicin-mediated cardiotoxicity via topoisomerase IIβ poisoning. Acute cardiac muscle damage occurs in response to the very first dose of doxorubicin, however, cardioprotection has been reported after co-treatment of doxorubicin with acyloxyalkyl ester prodrugs. The aim of this study was to examine the role played by various forms of acute cardiac damage mediated by doxorubicin and determine a mechanism for the cardioprotective effect of formaldehyde-releasing prodrug AN-9 (pivaloyloxymethyl butyrate). Doxorubicin-induced cardiac damage in BALB/c mice bearing mammary tumours was established with a single dose of doxorubicin (4 or 16 mg/kg) administered alone or in combination with AN-9 (100 mg/kg). AN-9 protected the heart from doxorubicin-induced myocardial apoptosis and also significantly reduced dsDNA breaks, independent from the level of doxorubicin biodistribution to the heart. Covalent incorporation of [14C]doxorubicin into DNA showed that the combination treatment yielded significantly higher levels of formaldehyde-mediated doxorubicin-DNA adducts compared to doxorubicin alone, yet this form of damage was associated with cardioprotection from apoptosis. The cardiac transcriptomic analysis indicates that the combination treatment initiates inflammatory response signalling pathways. Doxorubicin and AN-9 combination treatments were cardioprotective, yet preserved doxorubicin-mediated anti-tumour proliferation and apoptosis in mammary tumours. This was associated with a switch in doxorubicin action from cardiac topoisomerase IIβ poisoning to covalent-DNA adduct formation. Co-administration of doxorubicin and formaldehyde-releasing prodrugs, such as AN-9, may be a promising cardioprotective therapy while maintaining doxorubicin activity in primary mammary tumours.
Collapse
Affiliation(s)
- Alison Cheong
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Sean McGrath
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Tina Robinson
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Ruqaya Maliki
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Alex Spurling
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Peter Lock
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Ada Rephaeli
- Laboratory for Pharmacology and Experimental Oncology, Felsenstein Medical Research Center, Petach Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, 49100 Tel Aviv, Israel
| | - Abraham Nudelman
- Division of Medicinal Chemistry, Department of Chemistry, Bar-Ilan University, Ramat Gan 52900, Israel
| | - Belinda S Parker
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Salvatore Pepe
- Murdoch Children's Research Institute, Department of Cardiology, Royal Children's Hospital, Australia; Department of Paediatrics, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Suzanne M Cutts
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia.
| |
Collapse
|
17
|
Asiatic Acid Protects against Doxorubicin-Induced Cardiotoxicity in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:5347204. [PMID: 32509145 PMCID: PMC7246415 DOI: 10.1155/2020/5347204] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 01/28/2020] [Indexed: 11/18/2022]
Abstract
The use of doxorubicin (DOX) can result in depression of cardiac function and refractory cardiomyopathy. Currently, there are no effective approaches to prevent DOX-related cardiac complications. Asiatic acid (AA) has been reported to provide cardioprotection against several cardiovascular diseases. However, whether AA could attenuate DOX-related cardiac injury remains unclear. DOX (15 mg/kg) was injected intraperitoneally into the mice to mimic acute cardiac injury, and the mice were given AA (10 mg/kg or 30 mg/kg) for 2 weeks for protection. The data in our study found that AA-treated mice exhibited attenuated cardiac injury and improved cardiac function in response to DOX injection. AA also suppressed myocardial oxidative damage and apoptosis without affecting cardiac inflammation in DOX-treated mice. AA also provided protection in DOX-challenged cardiomyocytes, improved cell viability, and suppressed intracellular reactive oxygen species (ROS) in vitro. Detection of signaling pathways showed that AA activated protein kinase B (AKT) signaling pathway in vivo and in vitro. Furthermore, we found that AA lost its protective effects in the heart with AKT inactivation. In conclusion, our results found that AA could attenuate DOX-induced myocardial oxidative stress and apoptosis via activation of the AKT signaling pathway.
Collapse
|
18
|
Tetrandrine Attenuated Doxorubicin-Induced Acute Cardiac Injury in Mice. BIOMED RESEARCH INTERNATIONAL 2020; 2020:2616024. [PMID: 32461972 PMCID: PMC7232681 DOI: 10.1155/2020/2616024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 04/07/2020] [Accepted: 04/21/2020] [Indexed: 12/05/2022]
Abstract
Oxidative damage is closely involved in the development of doxorubicin- (DOX-) induced cardiotoxicity. It has been reported that tetrandrine can prevent the development of cardiac hypertrophy by suppressing reactive oxygen species- (ROS-) dependent signaling pathways in mice. However, whether tetrandrine could attenuate DOX-related cardiotoxicity remains unclear. To explore the protective effect of tetrandrine, mice were orally given a dose of tetrandrine (50 mg/kg) for 4 days beginning one day before DOX injection. To induce acute cardiac injury, the mice were exposed to a single intraperitoneal injection of DOX (15 mg/kg). The data in our study showed that tetrandrine prevented DOX-related whole-body wasting and heart atrophy, decreased markers of cardiac injury, and improved cardiac function in mice. Moreover, tetrandrine supplementation protected the mice against oxidative damage and myocardial apoptotic death. Tetrandrine supplementation also reduced ROS production and improved cell viability after DOX exposure in vitro. We also found that tetrandrine supplementation increased nuclear factor (erythroid-derived 2)-like 2 (Nrf2) expression and activity in vivo and in vitro. The protection of tetrandrine supplementation was blocked by Nrf2 deficiency in mice. In conclusion, our study found that tetrandrine could improve cardiac function and prevent the development of DOX-related cardiac injury through activation of Nrf2.
Collapse
|
19
|
Rahimi O, Kirby J, Varagic J, Westwood B, Tallant EA, Gallagher PE. Angiotensin-(1–7) reduces doxorubicin-induced cardiac dysfunction in male and female Sprague-Dawley rats through antioxidant mechanisms. Am J Physiol Heart Circ Physiol 2020; 318:H883-H894. [DOI: 10.1152/ajpheart.00224.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Doxorubicin (Dox) is an effective chemotherapeutic for a variety of pediatric malignancies. Unfortunately, Dox administration often results in a cumulative dose-dependent cardiotoxicity that manifests with marked oxidative stress, leading to heart failure. Adjunct therapies are needed to mitigate Dox cardiotoxicity and enhance quality of life in pediatric patients with cancer. Angiotensin-(1–7) [Ang-(1–7)] is an endogenous hormone with cardioprotective properties. This study investigated whether adjunct Ang-(1–7) attenuates cardiotoxicity resulting from exposure to Dox in male and female juvenile rats. Dox significantly reduced body mass, and the addition of Ang-(1–7) had no effect. However, adjunct Ang-(1–7) prevented Dox-mediated diastolic dysfunction, including markers of decreased passive filling as measured by reduced early diastole mitral valve flow velocity peak ( E) ( P < 0.05) and early diastole mitral valve annulus peak velocity ( e′; P < 0.001) and increased E/e′ ( P < 0.001), an echocardiographic measure of diastolic dysfunction. Since Dox treatment increases reactive oxygen species (ROS), the effect of Ang-(1–7) on oxidative by-products and enzymes that generate or reduce ROS was investigated. In hearts of male and female juvenile rats, Dox increased NADPH oxidase 4 ( P < 0.05), a major cardiovascular NADPH oxidase isozyme that generates ROS, as well as 4-hydroxynonenal ( P < 0.001) and malondialdehyde ( P < 0.001), markers of lipid peroxidation; Ang-(1–7) prevented these effects of Dox. Cotreatment with Dox and Ang-(1–7) increased the antioxidant enzymes SOD1 (male: P < 0.05; female: P < 0.01) and catalase ( P < 0.05), which likely contributed to reduced ROS. These results demonstrate that Ang-(1–7) prevents diastolic dysfunction in association with a reduction in ROS, suggesting that the heptapeptide hormone may serve as an effective adjuvant to improve Dox-induced cardiotoxicity. NEW & NOTEWORTHY Ang-(1–7) is a clinically safe peptide hormone with cardioprotective and antineoplastic properties that could be used as an adjuvant therapy to improve cancer treatment and mitigate the long-term cardiotoxicity associated with doxorubicin in pediatric patients with cancer.
Collapse
Affiliation(s)
- Omeed Rahimi
- Hypertension and Vascular Research, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Jay Kirby
- Hypertension and Vascular Research, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Jasmina Varagic
- Hypertension and Vascular Research, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Brian Westwood
- Hypertension and Vascular Research, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - E. Ann Tallant
- Hypertension and Vascular Research, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Patricia E. Gallagher
- Hypertension and Vascular Research, Wake Forest School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
20
|
Zhang H, Tian Y, Liang D, Fu Q, Jia L, Wu D, Zhu X. The Effects of Inhibition of MicroRNA-375 in a Mouse Model of Doxorubicin-Induced Cardiac Toxicity. Med Sci Monit 2020; 26:e920557. [PMID: 32186283 PMCID: PMC7102408 DOI: 10.12659/msm.920557] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Doxorubicin-induced myocardial toxicity is associated with oxidative stress, cardiomyocyte, apoptosis, and loss of contractile function. Previous studies showed that microRNA-375 (miR-375) expression was increased in mouse models of heart failure and clinically, and that inhibition of miR-375 reduced inflammation and increased survival of cardiomyocytes. This study aimed to investigate the effects and mechanisms of inhibition of miR-375 in a mouse model of doxorubicin-induced cardiac toxicity in vivo and in doxorubicin-treated rat and mouse cardiomyocytes in vitro. MATERIAL AND METHODS The mouse model of doxorubicin-induced cardiac toxicity was developed using an intraperitoneal injection of doxorubicin (15 mg/kg diluted in 0.9% saline) for eight days. Treatment was followed by a single subcutaneous injection of miR-375 inhibitor. H9c2 rat cardiac myocytes and adult murine cardiomyocytes (AMCs) were cultured in vitro and treated with doxorubicin, with and without pretreatment with miR-375 inhibitor. RESULTS Doxorubicin significantly upregulated miR-375 expression in vitro and in vivo, and inhibition of miR-375 re-established myocardial redox homeostasis, prevented doxorubicin-induced oxidative stress and cardiomyocyte apoptosis, and activated the PDK1/AKT axis by reducing the direct binding of miR-375 to 3' UTR of the PDK1 gene. Inhibition of PDK1 and AKT abolished the protective role of miR-375 inhibition on doxorubicin-induced oxidative damage. CONCLUSIONS Inhibition of miR-375 prevented oxidative damage in a mouse model of doxorubicin-induced cardiac toxicity in vivo and in doxorubicin-treated rat and mouse cardiomyocytes in vitro through the PDK1/AKT signaling pathway.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Cardiovascular Surgery, Tianjin Medical University General Hospital, Tianjing, China (mainland)
| | - Yikui Tian
- Department of Cardiovascular Surgery, Tianjin Medical University General Hospital, Tianjing, China (mainland)
| | - Degang Liang
- Department of Cardiovascular Surgery, Tianjin Medical University General Hospital, Tianjing, China (mainland)
| | - Qiang Fu
- Department of Cardiovascular Surgery, Tianjin Medical University General Hospital, Tianjing, China (mainland)
| | - Liqun Jia
- Department of Cardiovascular Surgery, Tianjin Medical University General Hospital, Tianjing, China (mainland)
| | - Dawei Wu
- Department of Cardiovascular Surgery, Tianjin Medical University General Hospital, Tianjing, China (mainland)
| | - Xinyuan Zhu
- Department of Cardiovascular Surgery, Tianjin Medical University General Hospital, Tianjing, China (mainland)
| |
Collapse
|
21
|
Li XQ, Liu YK, Yi J, Dong JS, Zhang PP, Wan L, Li K. MicroRNA-143 Increases Oxidative Stress and Myocardial Cell Apoptosis in a Mouse Model of Doxorubicin-Induced Cardiac Toxicity. Med Sci Monit 2020; 26:e920394. [PMID: 32170053 PMCID: PMC7085239 DOI: 10.12659/msm.920394] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 11/20/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Oxidative stress and myocardial apoptosis are features of doxorubicin-induced cardiac toxicity that can result in cardiac dysfunction. Previous studies showed that microRNA-143 (miR-143) was expressed in the myocardium and had a role in cardiac function. This study aimed to investigate the effects and possible molecular mechanisms of miR-143 on oxidative stress and myocardial cell apoptosis in a mouse model of doxorubicin-induced cardiac toxicity. MATERIAL AND METHODS Mice underwent intraperitoneal injection of doxorubicin (15 mg/kg) daily for eight days to develop the mouse model of doxorubicin-induced cardiac toxicity. Four days before doxorubicin administration, a group of mice was pretreated daily with a miR-143 antagonist (25 mg/kg/day) for four consecutive days by tail vein injection. The study included the use of a miR-143 antagomir, or anti-microRNA, an oligonucleotide that silenced endogenous microRNA (miR), and an agomir to miR-143, and also the AKT inhibitor, MK2206. Quantitative real-time polymerase chain reaction (qRT-PCR) and immunoblot analysis were used to measure mRNA and protein expression, respectively. RESULTS Doxorubicin treatment increased the expression of miR-143, which was reduced by the miR-143 antagomir. Overexpression of miR-143 increased doxorubicin-induced myocardial apoptosis and oxidative stress. The use of the miR-143 antagomir significantly activated protein kinase B (PKB) and AKT, which were reduced in the presence of the AKT inhibitor, MK2206. However, the use of the miR-143 antagomir further down-regulated AKT phosphorylation following doxorubicin treatment and increased AKT activation. CONCLUSIONS In a mouse model of doxorubicin-induced cardiac toxicity, miR-143 increased oxidative stress and myocardial cell apoptosis following doxorubicin treatment by inhibiting AKT.
Collapse
|
22
|
Boussada M, Ali RB, Chahbi A, Abdelkarim M, Fradj MKB, Dziri C, Bokri K, Akacha AB, El May MV. A new Thiocyanoacetamide protects rat sperm cells from Doxorubicin-triggered cytotoxicity whereas Selenium shows low efficacy: In vitro approach. Toxicol In Vitro 2019; 61:104587. [PMID: 31271807 DOI: 10.1016/j.tiv.2019.06.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 05/29/2019] [Accepted: 06/29/2019] [Indexed: 11/26/2022]
Abstract
Doxorubicin (DOX) exhibits a wide-ranging spectrum of antitumor activities which maintain its clinical use despite its devastating impact on highly proliferating cells. The present work was designed to develop a new approach which aims to protect male germ cells from DOX cytotoxicity. Thus, an assessment of the protective potential of a new thioamide analog (thiocyanoacetamide; TA) compared to selenium (Se) was performed in rat sperms exposed to DOX in vitro. Oxygen consumption rate (OCR) was measured after exposure to three different doses (0.5, 1, 1.5 and 2 μM) of DOX, Se or TA, and the suitable concentrations were selected for further studies afterwards. Motility, OCR in a time-dependent manner, glucose extracellular concentration and lipid peroxidation (LPO) were measured. Fatty acid (FA) content was assessed by gas chromatography (GC-FID). Cell death, superoxide anion (O2-), mitochondrial membrane potential (MMP), and DNA damage were evaluated by flow cytometry. TA association with DOX increased OCR and glucose uptake, improved cell survival and decreased DNA damage. The co-administration of DOX with Se increased OCR, significantly prevented O2- overproduction, and decreased LPO. Collected data brought new insights regarding this transformed TA, which showed better efficiency than Se in reducing DOX cytotoxic stress in sperms.
Collapse
Affiliation(s)
- Marwa Boussada
- UR17/ES/13 Laboratory of Histology and Embryology, Faculty of Medicine of Tunis, University of Tunis El Manar (UTM), Jabbari Jebel Lakhdar Street 15, 1007 Tunis, Tunisia.
| | - Ridha Ben Ali
- UR17/ES/13 Laboratory of Histology and Embryology, Faculty of Medicine of Tunis, University of Tunis El Manar (UTM), Jabbari Jebel Lakhdar Street 15, 1007 Tunis, Tunisia; Unity of Experimental Medicine, Faculty of Medicine of Tunis, University of Tunis El Manar (UTM), Jabbari Jebel Lakhdar Street 15, 1007 Tunis, Tunisia
| | - Ahlem Chahbi
- Laboratory of Hematology, Faculty of Medicine of Tunis, University of Tunis El Manar (UTM), Jabbari Jebel Lakhdar Street 15, 1007 Tunis, Tunisia.
| | - Mohamed Abdelkarim
- Laboratory of Hematology, Faculty of Medicine of Tunis, University of Tunis El Manar (UTM), Jabbari Jebel Lakhdar Street 15, 1007 Tunis, Tunisia
| | - Mohamed Kacem Ben Fradj
- UR05/08-08, LR99/ES/11, Department of Biochemistry, Rabta Hospital, University of Tunis El Manar (UTM), Jabbari Jebel Lakhdar Street 15, 1007 Tunis, Tunisia
| | - Chadli Dziri
- Unity of Experimental Medicine, Faculty of Medicine of Tunis, University of Tunis El Manar (UTM), Jabbari Jebel Lakhdar Street 15, 1007 Tunis, Tunisia
| | - Khouloud Bokri
- Laboratory of Organic Synthesis and Heterocyclic Chemistry Department, Faculty of Sciences of Tunis, University of Tunis El Manar, 2092 Tunis, Tunisia
| | - Azaiez Ben Akacha
- Laboratory of Organic Synthesis and Heterocyclic Chemistry Department, Faculty of Sciences of Tunis, University of Tunis El Manar, 2092 Tunis, Tunisia
| | - Michèle Véronique El May
- UR17/ES/13 Laboratory of Histology and Embryology, Faculty of Medicine of Tunis, University of Tunis El Manar (UTM), Jabbari Jebel Lakhdar Street 15, 1007 Tunis, Tunisia
| |
Collapse
|
23
|
miR-200a Attenuated Doxorubicin-Induced Cardiotoxicity through Upregulation of Nrf2 in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:1512326. [PMID: 31781322 PMCID: PMC6875222 DOI: 10.1155/2019/1512326] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 08/07/2019] [Accepted: 08/31/2019] [Indexed: 02/07/2023]
Abstract
Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) was closely involved in doxorubicin- (DOX-) induced cardiotoxicity. MicroRNA-200a (miR-200a) could target Keap1 mRNA and promote degradation of Keap1 mRNA, resulting in Nrf2 activation. However, the role of miR-200a in DOX-related cardiotoxicity remained unclear. Our study is aimed at investigating the effect of miR-200a on DOX-induced cardiotoxicity in mice. For cardiotropic expression, male mice received an injection of an adeno-associated virus 9 (AAV9) system carrying miR-200a or miR-scramble. Four weeks later, mice received a single intraperitoneal injection of DOX at 15 mg/kg. In our study, we found that miR-200a mRNA was the only microRNA that was significantly decreased in DOX-treated mice and H9c2 cells. miR-200a supplementation blocked whole-body wasting and heart atrophy caused by acute DOX injection, decreased the levels of cardiac troponin I and the N-terminal probrain natriuretic peptide, and improved cardiac and adult cardiomyocyte contractile function. Moreover, miR-200a reduced oxidative stress and cardiac apoptosis without affecting matrix metalloproteinase and inflammatory factors in mice with acute DOX injection. miR-200a also attenuated DOX-induced oxidative injury and cell loss in vitro. As expected, we found that miR-200a activated Nrf2 and Nrf2 deficiency abolished the protection provided by miR-200a supplementation in mice. miR-200a also provided cardiac benefits in a chronic model of DOX-induced cardiotoxicity. In conclusion, miR-200a protected against DOX-induced cardiotoxicity via activation of the Nrf2 signaling pathway. Our data suggest that miR-200a may represent a new cardioprotective strategy against DOX-induced cardiotoxicity.
Collapse
|
24
|
Xu C, Liu CH, Zhang DL. MicroRNA-22 inhibition prevents doxorubicin-induced cardiotoxicity via upregulating SIRT1. Biochem Biophys Res Commun 2019; 521:485-491. [PMID: 31677784 DOI: 10.1016/j.bbrc.2019.10.140] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 10/19/2019] [Indexed: 01/09/2023]
Abstract
Oxidative stress and cardiomyocyte apoptosis contributed to the progression of doxorubicin (Dox)-induced cardiotoxicity. Recent studies identified microRNA-22 (miR-22) as a cardiac- and skeletal muscle-enriched microRNA that functioned as a key regulator in stress-induced cardiac injury. The present study aimed to investigate the role and possible mechanism of miR-22 on Dox-induced oxidative stress and cardiomyocyte apoptosis. Mice were exposed to reduplicative injections of Dox (i.p., 4 mg/kg) weekly for consecutive 4 weeks to generate Dox-induced cardiotoxicity. Herein, we found that miR-22 level was significantly increased in murine hearts subjected to chronic Dox treatment. MiR-22 inhibition attenuated oxidative stress and cardiomyocyte apoptosis in vivo and in vitro, thereby preventing Dox-induced cardiac dysfunction. Mechanistically, we observed that miR-22 directly bound to the 3'-UTR of Sirt1 and caused SIRT1 downregulation. Conversely, miR-22 antagomir upregulated SIRT1 expression and SIRT1 inhibitor abolished the beneficial effects of miR-22 antagomir. In conclusion, miR-22 inhibition prevented oxidative stress and cardiomyocyte apoptosis via upregulating SIRT1 and miR-22 might be a new target for treating Dox-induced cardiotoxicity.
Collapse
Affiliation(s)
- Can Xu
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang, Hunan, 421001, PR China
| | - Chang-Hui Liu
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang, Hunan, 421001, PR China
| | - Da-Li Zhang
- Department of Emergency, The First Affiliated Hospital of University of South China, Hengyang, Hunan, 421001, PR China.
| |
Collapse
|
25
|
miR-451 Silencing Inhibited Doxorubicin Exposure-Induced Cardiotoxicity in Mice. BIOMED RESEARCH INTERNATIONAL 2019; 2019:1528278. [PMID: 31355248 PMCID: PMC6637715 DOI: 10.1155/2019/1528278] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 06/10/2019] [Indexed: 12/15/2022]
Abstract
Oxidative stress and cardiomyocytes apoptosis were closely involved in the pathological process of doxorubicin- (Dox-) induced cardiac injury. MicroRNA-451 (miR-451) was mainly expressed in cardiomyocytes. However, the role of miR-451 in Dox-induced cardiac injury remained unclear. Our study aimed to investigate the effect of miR-451 on Dox-induced cardiotoxicity in mice. We established a Dox-induced cardiotoxicity model in the mice and manipulated miR-451 expression in the heart using a miR-451 inhibitor, which was injected every other day beginning at one day before Dox injection. Oxidative stress and apoptosis in the hearts were evaluated. miR-451 levels were significantly increased in Dox-treated mice or cardiomyocytes. miR-451 inhibition attenuated Dox-induced whole-body wasting and heart atrophy, reduced cardiac injury, restored cardiac function, and improved cardiomyocyte contractile function. Moreover, miR-451 inhibition reduced oxidative stress and cardiomyocytes apoptosis in vivo and in vitro. miR-451 inhibition increased the expression of calcium binding protein 39 (Cab39) and activated adenosine monophosphate activated protein kinase (AMPK) signaling pathway. A specific inhibitor of AMPK abolished the protection provided by miR-451 inhibition against cell injury in vitro. In conclusion, miR-451 inhibition protected against Dox-induced cardiotoxicity via activation of AMPK signaling pathway.
Collapse
|
26
|
Yarana C, Thompson H, Chaiswing L, Butterfield DA, Weiss H, Bondada S, Alhakeem S, Sukati S, St Clair DK. Extracellular vesicle-mediated macrophage activation: An insight into the mechanism of thioredoxin-mediated immune activation. Redox Biol 2019; 26:101237. [PMID: 31276937 PMCID: PMC6612011 DOI: 10.1016/j.redox.2019.101237] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 05/28/2019] [Accepted: 05/31/2019] [Indexed: 12/21/2022] Open
Abstract
Extracellular vesicles (EVs) generated from redox active anticancer drugs are released into the extracellular environment. These EVs contain oxidized molecules and trigger inflammatory responses by macrophages. Using a mouse model of doxorubicin (DOX)-induced tissue injury, we previously found that the major sources of circulating EVs are from heart and liver, organs that are differentially affected by DOX. Here, we investigated the effects of EVs from cardiomyocytes and those from hepatocytes on macrophage activation. EVs from H9c2 rat cardiomyocytes (H9c2 EVs) and EVs from FL83b mouse hepatocytes (FL83 b EVs) have different levels of protein-bound 4-hydroxynonenal and thus different immunostimulatory effects on mouse RAW264.7 macrophages. H9c2 EVs but not FL83 b EVs induced both pro-inflammatory and anti-inflammatory macrophage activation, mediated by NFκB and Nrf-2 pathways, respectively. DOX enhanced the effects of H9c2 EVs but not FL83 b EVs. While EVs from DOX-treated H9c2 cells (H9c2 DOXEVs) suppressed mitochondrial respiration and increased glycolysis of macrophages, EVs from DOX-treated FL83b cells (FL83b DOXEVs) enhanced mitochondrial reserve capacity. Mechanistically, the different immunostimulatory functions of H9c2 EVs and FL83 b EVs are regulated, in part, by the redox status of the cytoplasmic thioredoxin 1 (Trx1) of macrophages. H9c2 DOXEVs lowered the level of reduced Trx1 in cytoplasm while FL83b DOXEVs did the opposite. Trx1 overexpression alleviated the effect of H9c2 DOXEVs on NFκB and Nrf-2 activation and prevented the upregulation of their target genes. Our findings identify EVs as a novel Trx1-mediated redox mediator of immune response, which greatly enhances our understanding of innate immune responses during cancer therapy.
Collapse
Affiliation(s)
- Chontida Yarana
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Salaya, 73170, Thailand
| | - Hannah Thompson
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, 40536, USA
| | - Luksana Chaiswing
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, 40536, USA
| | - D Allan Butterfield
- Markey Cancer Center, University of Kentucky, Lexington, KY, 40536, USA; Department of Chemistry, University of Kentucky, Lexington, KY, 40506, USA
| | - Heidi Weiss
- Markey Cancer Center, University of Kentucky, Lexington, KY, 40536, USA
| | - Subbarao Bondada
- Department of Microbiology, Immunology & Molecular Genetics, University of Kentucky, Lexington, KY, 40536, USA
| | - Sara Alhakeem
- Department of Microbiology, Immunology & Molecular Genetics, University of Kentucky, Lexington, KY, 40536, USA
| | - Suriyan Sukati
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, 40536, USA; School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat, 80161, Thailand
| | - Daret K St Clair
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, 40536, USA.
| |
Collapse
|
27
|
Shabalala SC, Dludla PV, Muller CJF, Nxele X, Kappo AP, Louw J, Johnson R. Aspalathin ameliorates doxorubicin-induced oxidative stress in H9c2 cardiomyoblasts. Toxicol In Vitro 2018; 55:134-139. [PMID: 30576852 DOI: 10.1016/j.tiv.2018.12.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 11/28/2018] [Accepted: 12/18/2018] [Indexed: 10/27/2022]
Abstract
Aspalathin (ASP) is a C-dihydrochalcone abundantly found in Aspalathus linearis. While we have provide evidence that ASP can protect H9c2 cardiomyoblasts against doxorubicin (Dox)-induced apoptosis through regulation of autophagy, the complete mechanism involved in the cardioprotective effect of this dihydrochalcone remains to be explored. Here we provide evidence that ASP reverses Dox-induced apoptosis through the amelioration of oxidative stress in H9c2 cardiomyoblasts. Cultured cells were treated with 0.2 μM Dox or co-treated with either 20 μM dexrazoxane (Dexra) or 0.2 μM ASP daily for five days, to a final dose of 1 μM Dox, 100 μM Dexra and 1 μM ASP, respectively. Superoxide dismutase, catalase, glutathione, malondialdehyde and dichloro-dihydro-fluorescein diacetate fluorescence were used as end-point measurements for oxidative stress, while JC-1 and TUNEL labeling were performed to assess mitochondria depolarization and apoptosis. Co-treatment with ASP attenuated Dox-induced cardiotoxicity by improving endogenous antioxidant levels and mitochondrial membrane potential, while inhibiting reactive oxygen species production and cellular apoptosis. These findings suggested that ASP can prevent Dox-induced oxidative stress and apoptosis and needs further assessment to confirm its therapeutic potential to prevent Dox-induced cardiotoxicity.
Collapse
Affiliation(s)
- Samukelisiwe C Shabalala
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council (SAMRC), Tygerberg 7505, South Africa; Department of Biochemistry and Microbiology, Faculty of Science and Agriculture, University of Zululand, Kwadlangezwa 3886, South Africa
| | - Phiwayinkosi V Dludla
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council (SAMRC), Tygerberg 7505, South Africa
| | - Christo J F Muller
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council (SAMRC), Tygerberg 7505, South Africa; Department of Biochemistry and Microbiology, Faculty of Science and Agriculture, University of Zululand, Kwadlangezwa 3886, South Africa; Department of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg 7505, South Africa
| | - Xolisa Nxele
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council (SAMRC), Tygerberg 7505, South Africa; Department of Biotechnology, Faculty of Natural Sciences, University of the Western Cape, Bellville 7535, South Africa
| | - Abidemi P Kappo
- Department of Biochemistry and Microbiology, Faculty of Science and Agriculture, University of Zululand, Kwadlangezwa 3886, South Africa
| | - Johan Louw
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council (SAMRC), Tygerberg 7505, South Africa; Department of Biochemistry and Microbiology, Faculty of Science and Agriculture, University of Zululand, Kwadlangezwa 3886, South Africa
| | - Rabia Johnson
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council (SAMRC), Tygerberg 7505, South Africa; Department of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg 7505, South Africa.
| |
Collapse
|
28
|
Inhibition of vascular smooth muscle cells premature senescence with rutin attenuates and stabilizes diabetic atherosclerosis. J Nutr Biochem 2018; 51:91-98. [DOI: 10.1016/j.jnutbio.2017.09.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 08/07/2017] [Accepted: 09/12/2017] [Indexed: 01/21/2023]
|
29
|
Wen SY, Tsai CY, Pai PY, Chen YW, Yang YC, Aneja R, Huang CY, Kuo WW. Diallyl trisulfide suppresses doxorubicin-induced cardiomyocyte apoptosis by inhibiting MAPK/NF-κB signaling through attenuation of ROS generation. ENVIRONMENTAL TOXICOLOGY 2018; 33:93-103. [PMID: 29087013 DOI: 10.1002/tox.22500] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 09/29/2017] [Accepted: 10/06/2017] [Indexed: 06/07/2023]
Abstract
BACKGROUND Doxorubicin (Dox) is an effective anticancer agent. However, its effectiveness is limited by its cardiotoxic effects. It has also been reported that the mitogen-activated protein kinase family and NF-κB can be activated by Dox treatment. DATS has been shown to be a potent antioxidant with cardioprotective effects. We investigate whether Dox induces cardiac apoptosis through JNK- and ERK-dependent NF-κB upregulation that can be reduced by DATS treatment. METHODS AND MATERIAL H9c2 cells were treated with 0.5-1.5 μM Dox for 24 hours. Dox promoted apoptosis and ROS generation and inhibited viability in a dose-dependent manner. Then, the phosphorylation levels of JNK, ERK, and NF-κB evaluated by western blot were elevated. We used inhibitors of JNK, ERK, and NF-κB to determine which of these proteins were involved in Dox-induced apoptosis. Furthermore, Dox-exposed cells were treated with DATS at doses of 1, 5, and 10 μM, and the data demonstrated that ROS generation and apoptotic proteins were decreased and that ERK and NF-κB were downregulated in a dose-dependent manner. Additionally, six-week-old rats were divided into three groups (n = 6 per group) designed as an eight-week study. Normal, Dox (at dose 3.75 mg/kg by ip) administered with or without DATS (at dose 40 mg/kg by gavage) treatment groups. The results indicate that cardiac dysfunction, apoptosis, and JNK, ERK, and NF-κB activation by Dox were reversed by treatment with DATS. CONCLUSION DATS appears to suppress Dox-induced cardiomyocyte apoptosis by inhibiting NADPH oxidase-related ROS production and the downstream JNK/ERK/NF-κB signaling pathway; DATS may possess clinical therapeutic potential by blocking Dox-induced cardiotoxicity.
Collapse
Affiliation(s)
- Su-Ying Wen
- Department of Dermatology, Taipei City Hospital, Renai Branch, Taipei, Taiwan
- Center for General Education, Mackay Junior College of Medicine, Nursing, and Management, Taipei, Taiwan
| | - Cheng-Yen Tsai
- Department of Pediatrics, China Medical University Beigang Hospital, Yunlin, Taiwan, 651, Republic of China
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan, 404, Republic of China
| | - Pei-Ying Pai
- Department of Pathology, China Medical University Hospital, Taichung, Taiwan
| | - Yi-Wei Chen
- Department of Biological Science and Technology, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, Taiwan, 404, Republic of China
| | - Yao-Chih Yang
- Department of Biological Science and Technology, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, Taiwan, 404, Republic of China
| | - Ritu Aneja
- Department of Biology, Georgia State University, Atlanta, Georgia, 30303
| | - Chih-Yang Huang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan, 404, Republic of China
- Department of Chinese Medicine, China Medical University Hospital, Taichung, Taiwan, 404, Republic of China
- Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan, 413, Republic of China
| | - Wei-Wen Kuo
- Department of Biological Science and Technology, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, Taiwan, 404, Republic of China
| |
Collapse
|
30
|
Yarana C, Carroll D, Chen J, Chaiswing L, Zhao Y, Noel T, Alstott M, Bae Y, Dressler EV, Moscow JA, Butterfield DA, Zhu H, St Clair DK. Extracellular Vesicles Released by Cardiomyocytes in a Doxorubicin-Induced Cardiac Injury Mouse Model Contain Protein Biomarkers of Early Cardiac Injury. Clin Cancer Res 2017; 24:1644-1653. [PMID: 29070527 DOI: 10.1158/1078-0432.ccr-17-2046] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 09/08/2017] [Accepted: 10/18/2017] [Indexed: 01/21/2023]
Abstract
Purpose: Cardiac injury is a major cause of death in cancer survivors, and biomarkers for it are detectable only after tissue injury has occurred. Extracellular vesicles (EV) remove toxic biomolecules from tissues and can be detected in the blood. Here, we evaluate the potential of using circulating EVs as early diagnostic markers for long-term cardiac injury.Experimental Design: Using a mouse model of doxorubicin (DOX)-induced cardiac injury, we quantified serum EVs, analyzed proteomes, measured oxidized protein levels in serum EVs released after DOX treatment, and investigated the alteration of EV content.Results: Treatment with DOX caused a significant increase in circulating EVs (DOX_EV) compared with saline-treated controls. DOX_EVs exhibited a higher level of 4-hydroxynonenal adducted proteins, a lipid peroxidation product linked to DOX-induced cardiotoxicity. Proteomic profiling of DOX_EVs revealed the distinctive presence of brain/heart, muscle, and liver isoforms of glycogen phosphorylase (GP), and their origins were verified to be heart, skeletal muscle, and liver, respectively. The presence of brain/heart GP (PYGB) in DOX_EVs correlated with a reduction of PYGB in heart, but not brain tissues. Manganese superoxide dismutase (MnSOD) overexpression, as well as pretreatment with cardioprotective agents and MnSOD mimetics, resulted in a reduction of EV-associated PYGB in mice treated with DOX. Kinetic studies indicated that EVs containing PYGB were released prior to the rise of cardiac troponin in the blood after DOX treatment, suggesting that PYGB is an early indicator of cardiac injury.Conclusions: EVs containing PYGB are an early and sensitive biomarker of cardiac injury. Clin Cancer Res; 24(7); 1644-53. ©2017 AACRSee related commentary by Zhu and Gius, p. 1516.
Collapse
Affiliation(s)
- Chontida Yarana
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky.,Faculty of Medical Technology, Mahidol University, Salaya, Thailand
| | - Dustin Carroll
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky
| | - Jing Chen
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky
| | - Luksana Chaiswing
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky
| | - Yanming Zhao
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky
| | - Teresa Noel
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky
| | - Michael Alstott
- Markey Cancer Center, Redox Metabolism Shared Resource Facility, University of Kentucky, Lexington, Kentucky
| | - Younsoo Bae
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky
| | - Emily V Dressler
- Division of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Jeffrey A Moscow
- Investigational Drug Branch, National Cancer Institute, Bethesda, Maryland
| | - D Allan Butterfield
- Markey Cancer Center, Redox Metabolism Shared Resource Facility, University of Kentucky, Lexington, Kentucky.,Department of Chemistry, University of Kentucky, Lexington, Kentucky
| | - Haining Zhu
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky.,Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky.,Markey Cancer Center, Redox Metabolism Shared Resource Facility, University of Kentucky, Lexington, Kentucky
| | - Daret K St Clair
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky.
| |
Collapse
|
31
|
Xu F, Li X, Liu L, Xiao X, Zhang L, Zhang S, Lin P, Wang X, Wang Y, Li Q. Attenuation of doxorubicin-induced cardiotoxicity by esculetin through modulation of Bmi-1 expression. Exp Ther Med 2017; 14:2216-2220. [PMID: 28962145 PMCID: PMC5609155 DOI: 10.3892/etm.2017.4763] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 07/04/2017] [Indexed: 01/27/2023] Open
Abstract
The protective effects and mechanisms of esculetin on doxorubicin (DOX)-induced injury of H9c2 cells were investigated. H9c2 cells were cultured and the logarithmic growth phase of the cells was divided into a control group, a DOX group and an esculetin + DOX group. Cell viability was detected by MTT assay. Annexin V-PI (AV-PI) double staining flow cytometry was carried out to detect cell apoptosis. Intracellular reactive oxygen species (ROS) were detected by flow cytometry. Transmission electron microscope (TEM) was used to evaluate cell ultrastructure. Cleaved caspase-3, cleaved PARP, Bcl-2, Bid and Bmi-1 proteins levels were investigated by western blot analysis. Bmi-1 siRNA was used to detect the role of Bmi-1 in the protective effects of esculetin against DOX-induced toxicity in H9c2 cells. The MTT and AV-PI double staining results showed that esculetin significantly increased H9c2 cell viability. Compared with the control group, the levels of cleaved caspase-3, cleaved PARP, Bid and ROS levels were significantly decreased, but the expression of Bcl-2 and Bmi-1 were significantly increased in the esculetin + DOX group. TEM showed that the cell structure of the mitochondria was protected by esculetin. The results of Bmi-1 siRNA showed that esculetin could protect DOX-induced cardiotoxicity by modulating Bmi-1 expression. Esculetin can protect DOX-induced cardiotoxicity and the effects may be attributable to modulation of Bmi-1 expression, provoking intracellular ROS accumulation, protecting the structure of mitochondria and reducing cell apoptosis.
Collapse
Affiliation(s)
- Fan Xu
- Department of Oncology, Affiliated Hospital of Chengde Medical College, Chengde, Hebei 067000, P.R. China
| | - Xiao Li
- Department of Radiology, Affiliated Hospital of Chengde Medical College, Chengde, Hebei 067000, P.R. China
| | - Lanfang Liu
- Department of Oncology, Affiliated Hospital of Chengde Medical College, Chengde, Hebei 067000, P.R. China
| | - Xu Xiao
- Department of Pharmacy, Affiliated Hospital of Chengde Medical College, Chengde, Hebei 067000, P.R. China
| | - Li Zhang
- Department of Oncology, Affiliated Hospital of Chengde Medical College, Chengde, Hebei 067000, P.R. China
| | - Shenglin Zhang
- Department of Oncology, Affiliated Hospital of Chengde Medical College, Chengde, Hebei 067000, P.R. China
| | - Pingping Lin
- Department of Oncology, Affiliated Hospital of Chengde Medical College, Chengde, Hebei 067000, P.R. China
| | - Xiaojie Wang
- Basic Medical Institute, Chengde Medical College, Chengde, Hebei 067000, P.R. China
| | - Yongwei Wang
- Department of Anatomy, Chengde Medical College, Chengde, Hebei 067000, P.R. China
| | - Qingshan Li
- Department of Oncology, Affiliated Hospital of Chengde Medical College, Chengde, Hebei 067000, P.R. China
| |
Collapse
|
32
|
Ramdath DD, Padhi EMT, Sarfaraz S, Renwick S, Duncan AM. Beyond the Cholesterol-Lowering Effect of Soy Protein: A Review of the Effects of Dietary Soy and Its Constituents on Risk Factors for Cardiovascular Disease. Nutrients 2017; 9:E324. [PMID: 28338639 PMCID: PMC5409663 DOI: 10.3390/nu9040324] [Citation(s) in RCA: 151] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Revised: 03/16/2017] [Accepted: 03/21/2017] [Indexed: 12/13/2022] Open
Abstract
The hypocholesterolemic effect of soy is well-documented and this has led to the regulatory approval of a health claim relating soy protein to a reduced risk of cardiovascular disease (CVD). However, soybeans contain additional components, such as isoflavones, lecithins, saponins and fiber that may improve cardiovascular health through independent mechanisms. This review summarizes the evidence on the cardiovascular benefits of non-protein soy components in relation to known CVD risk factors such as hypertension, hyperglycemia, inflammation, and obesity beyond cholesterol lowering. Overall, the available evidence suggests non-protein soy constituents improve markers of cardiovascular health; however, additional carefully designed studies are required to independently elucidate these effects. Further, work is also needed to clarify the role of isoflavone-metabolizing phenotype and gut microbiota composition on biological effect.
Collapse
Affiliation(s)
- D Dan Ramdath
- Guelph Research and Development Centre, Agriculture and Agri-Food Canada, Guelph, ON N1G 5C9, Canada.
| | - Emily M T Padhi
- Guelph Research and Development Centre, Agriculture and Agri-Food Canada, Guelph, ON N1G 5C9, Canada.
| | - Sidra Sarfaraz
- Guelph Research and Development Centre, Agriculture and Agri-Food Canada, Guelph, ON N1G 5C9, Canada.
| | - Simone Renwick
- Guelph Research and Development Centre, Agriculture and Agri-Food Canada, Guelph, ON N1G 5C9, Canada.
| | - Alison M Duncan
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON N1G 2E1, Canada.
| |
Collapse
|
33
|
Repka CP, Hayward R. Oxidative Stress and Fitness Changes in Cancer Patients after Exercise Training. Med Sci Sports Exerc 2016; 48:607-14. [PMID: 26587845 DOI: 10.1249/mss.0000000000000821] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
INTRODUCTION The purpose of this study was to determine the effect of an exercise intervention (EX) on muscular strength, cardiorespiratory fitness (CRF), and oxidative stress in cancer survivors compared with a nonexercising cancer control group (CON). METHODS Fifteen cancer patients and seven age-matched individuals with no history of cancer (NC) participated in this study. A blood draw and assessments of muscular strength and CRF were administered to cancer survivors within 6 wk of completing radiation or chemotherapy, and again 10 wk later. Eight cancer patients completed a 10-wk supervised exercise intervention, whereas seven continued standard care. Baseline oxidative stress was compared between cancer patients and the NC group. Changes in plasma protein carbonyls, 8-OHdG, and Trolox equivalent antioxidant capacity were compared between groups using repeated-measures ANOVA, and correlations between fitness and oxidative stress changes were evaluated. RESULTS Baseline antioxidant capacity was significantly lower, and plasma protein carbonyls were significantly higher in cancer patients compared with NC (P = 0.001). EX had a significant increase in antioxidant capacity (P < 0.001) and decrease in protein carbonyls (P = 0.023), whereas CON did not. Improvements in composite arm (41%, P = 0.002) and leg strength (34%, P = 0.008), isometric handgrip strength (11%, P = 0.015), and V˙O2peak (16%, P = 0.018) were significant in EX but not in CON. 8-OHdG changes were significantly correlated with changes in V˙O2peak (r = -0.89, P < 0.001), arm strength (r = -0.67, P = 0.004), and leg strength (r = -0.56, P = 0.019). CONCLUSION A whole-body exercise intervention for cancer survivors may be an effective method of concurrently increasing muscular strength, CRF, and antioxidant capacity while decreasing markers of oxidative stress.
Collapse
Affiliation(s)
- Chris P Repka
- 1Department of Health Sciences, Northern Arizona University, Flagstaff, AZ; 2School of Sport and Exercise Science and the Rocky Mountain Cancer Rehabilitation Institute, University of Northern Colorado, Greeley, CO
| | | |
Collapse
|
34
|
Abstract
Doxorubicin is the highly effective anthracycline, but its clinical use is limited by cardiotoxicity and consequent dysfunction. It has been proposed that the etiology of this is related to mitochondrial dysfunction. Connexin 43 (Cx43), the principal protein building block of cardiac gap junctions and hemichannels, plays an important role in cardioprotection. Recent reports confirmed the presence of Cx43 in the mitochondria as well. In this study, the role of mitochondrial Cx43 was evaluated 3 or 6 h after Doxorubicin administration to the rat heart cell line H9c2. Pharmacological inhibition of Hsp90 demonstrated that the mitochondrial Cx43 conferred cardioprotection by reducing cytosolic and mitochondrial reactive oxygen species production, mitochondrial calcium overload and mitochondrial membrane depolarization and cytochrome c release. In conclusion, our study demonstrates that Cx43 plays an important role in the protection of cardiac cells from Doxorubicin-induced toxicity.
Collapse
|
35
|
Dębowska K, Dębski D, Michałowski B, Dybala-Defratyka A, Wójcik T, Michalski R, Jakubowska M, Selmi A, Smulik R, Piotrowski Ł, Adamus J, Marcinek A, Chlopicki S, Sikora A. Characterization of Fluorescein-Based Monoboronate Probe and Its Application to the Detection of Peroxynitrite in Endothelial Cells Treated with Doxorubicin. Chem Res Toxicol 2016; 29:735-46. [PMID: 27081868 DOI: 10.1021/acs.chemrestox.5b00431] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Boronate probes have emerged recently as a versatile tool for the detection of reactive oxygen and nitrogen species. Here, we present the characterization of a fluorescein-based monoboronate probe, a 4-(pinacol boronate)benzyl derivative of fluorescein methyl ester (FBBE), that proved to be useful to detect peroxynitrite in cell culture experiments. The reactivity of FBBE toward peroxynitrite as well hypochlorite, hydrogen peroxide, and tyrosyl hydroperoxide was determined. Second-order rate constants of the reactions of FBBE with peroxynitrite, HOCl, and H2O2 at pH 7.4 were equal to (2.8 ± 0.2) × 10(5) M(-1) s(-1), (8.6 ± 0.5) × 10(3) M(-1) s(-1), and (0.96 ± 0.03) M(-1) s(-1), respectively. The presence of glutathione completely blocked the oxidation of the probe by HOCl and significantly inhibited its oxidation by H2O2 and tyrosyl hydroperoxide but not by peroxynitrite. The oxidative conversion of the probe was also studied in the systems generating singlet oxygen, superoxide radical anion, and nitric oxide in the presence and absence of glutathione. Spectroscopic characterization of FBBE and its oxidation product has been also performed. The differences in the reactivity pattern were supported by DFT quantum mechanical calculations. Finally, the FBBE probe was used to study the oxidative stress in endothelial cells (Ea.hy926) incubated with doxorubicin, a quinone anthracycline antibiotic. In endothelial cells pretreated with doxorubicin, FBBE was oxidized, and this effect was reversed by PEG-SOD and L-NAME but not by catalase.
Collapse
Affiliation(s)
- Karolina Dębowska
- Institute of Applied Radiation Chemistry, Lodz University of Technology , Lodz, Poland
| | - Dawid Dębski
- Institute of Applied Radiation Chemistry, Lodz University of Technology , Lodz, Poland
| | - Bartosz Michałowski
- Institute of Applied Radiation Chemistry, Lodz University of Technology , Lodz, Poland
| | | | - Tomasz Wójcik
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University , Kraków, Poland
| | - Radosław Michalski
- Institute of Applied Radiation Chemistry, Lodz University of Technology , Lodz, Poland
| | - Małgorzata Jakubowska
- Institute of Applied Radiation Chemistry, Lodz University of Technology , Lodz, Poland
| | - Anna Selmi
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University , Kraków, Poland
| | - Renata Smulik
- Institute of Applied Radiation Chemistry, Lodz University of Technology , Lodz, Poland
| | - Łukasz Piotrowski
- Institute of Applied Radiation Chemistry, Lodz University of Technology , Lodz, Poland
| | - Jan Adamus
- Institute of Applied Radiation Chemistry, Lodz University of Technology , Lodz, Poland
| | - Andrzej Marcinek
- Institute of Applied Radiation Chemistry, Lodz University of Technology , Lodz, Poland
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University , Kraków, Poland.,Chair of Pharmacology, Jagiellonian University Medical College , Kraków, Poland
| | - Adam Sikora
- Institute of Applied Radiation Chemistry, Lodz University of Technology , Lodz, Poland
| |
Collapse
|
36
|
Zhang W, Deng J, Sunkara M, Morris AJ, Wang C, St Clair D, Vore M. Loss of multidrug resistance-associated protein 1 potentiates chronic doxorubicin-induced cardiac dysfunction in mice. J Pharmacol Exp Ther 2015; 355:280-7. [PMID: 26354995 PMCID: PMC4613956 DOI: 10.1124/jpet.115.225581] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 08/26/2015] [Indexed: 12/20/2022] Open
Abstract
Doxorubicin (DOX), an effective cancer chemotherapeutic agent, induces dose-dependent cardiotoxicity, in part due to its ability to cause oxidative stress. We investigated the role of multidrug resistance-associated protein 1 (Mrp1/Abcc1) in DOX-induced cardiotoxicity in C57BL wild-type (WT) mice and their Mrp1 null (Mrp1(-/-)) littermates. Male mice were administered intraperitoneal DOX (3 or 2 mg/kg body weight) or saline twice a week for 3 weeks and examined 2 weeks after the last dose (protocol A total dose: 18 mg/kg) or for 5 weeks, and mice were examined 48 hours and 2 weeks after the last dose (protocol B total dose: 20 mg/kg). Chronic DOX induced body weight loss and hemotoxicity, adverse effects significantly exacerbated in Mrp1(-/-) versus WT mice. In the heart, significantly higher basal levels of glutathione (1.41-fold ± 0.27-fold) and glutathione disulfide (1.35-fold ± 0.16-fold) were detected in Mrp1(-/-) versus WT mice, and there were comparable decreases in the glutathione/glutathione disulfide ratio in WT and Mrp1(-/-) mice after DOX administration. Surprisingly, DOX induced comparable increases in 4-hydroxynonenal glutathione conjugate concentration in hearts from WT and Mrp1(-/-) mice. However, more DOX-induced apoptosis was detected in Mrp1(-/-) versus WT hearts (P < 0.05) (protocol A), and cardiac function, assessed by measurement of fractional shortening and ejection fraction with echocardiography, was significantly decreased by DOX in Mrp1(-/-) versus WT mice (P < 0.05; 95% confidence intervals of 20.0%-24.3% versus 23.7%-29.5% for fractional shortening, and 41.5%-48.4% versus 47.7%-56.7% for ejection fraction; protocol B). Together, these data indicate that Mrp1 protects the mouse heart against chronic DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Toxicology and Cancer Biology (W.Z., J.D., D.S.C., M.V.), Division of Cardiovascular Medicine, (M.S., A.J.M), and Markey Cancer Center (C.W.), College of Medicine, University of Kentucky, Lexington, Kentucky
| | - Jun Deng
- Department of Toxicology and Cancer Biology (W.Z., J.D., D.S.C., M.V.), Division of Cardiovascular Medicine, (M.S., A.J.M), and Markey Cancer Center (C.W.), College of Medicine, University of Kentucky, Lexington, Kentucky
| | - Manjula Sunkara
- Department of Toxicology and Cancer Biology (W.Z., J.D., D.S.C., M.V.), Division of Cardiovascular Medicine, (M.S., A.J.M), and Markey Cancer Center (C.W.), College of Medicine, University of Kentucky, Lexington, Kentucky
| | - Andrew J Morris
- Department of Toxicology and Cancer Biology (W.Z., J.D., D.S.C., M.V.), Division of Cardiovascular Medicine, (M.S., A.J.M), and Markey Cancer Center (C.W.), College of Medicine, University of Kentucky, Lexington, Kentucky
| | - Chi Wang
- Department of Toxicology and Cancer Biology (W.Z., J.D., D.S.C., M.V.), Division of Cardiovascular Medicine, (M.S., A.J.M), and Markey Cancer Center (C.W.), College of Medicine, University of Kentucky, Lexington, Kentucky
| | - Daret St Clair
- Department of Toxicology and Cancer Biology (W.Z., J.D., D.S.C., M.V.), Division of Cardiovascular Medicine, (M.S., A.J.M), and Markey Cancer Center (C.W.), College of Medicine, University of Kentucky, Lexington, Kentucky
| | - Mary Vore
- Department of Toxicology and Cancer Biology (W.Z., J.D., D.S.C., M.V.), Division of Cardiovascular Medicine, (M.S., A.J.M), and Markey Cancer Center (C.W.), College of Medicine, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
37
|
Deng J, Coy D, Zhang W, Sunkara M, Morris AJ, Wang C, Chaiswing L, St Clair D, Vore M, Jungsuwadee P. Elevated glutathione is not sufficient to protect against doxorubicin-induced nuclear damage in heart in multidrug resistance-associated protein 1 (Mrp1/Abcc1) null mice. J Pharmacol Exp Ther 2015; 355:272-9. [PMID: 26354996 PMCID: PMC4613962 DOI: 10.1124/jpet.115.225490] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 08/26/2015] [Indexed: 12/18/2022] Open
Abstract
Cardiotoxicity is a major dose-limiting adverse effect of doxorubicin (DOX), mediated in part by overproduction of reactive oxygen species and oxidative stress. Abcc1 (Mrp1) mediates the efflux of reduced and oxidized glutathione (GSH, GSSG) and is also a major transporter that effluxes the GSH conjugate of 4-hydroxy-2-nonenal (HNE; GS-HNE), a toxic product of lipid peroxidation formed during oxidative stress. To assess the role of Mrp1 in protecting the heart from DOX-induced cardiac injury, wild-type (WT) and Mrp1 null (Mrp1(-/-)) C57BL/6 littermate mice were administered DOX (15 mg/kg) or saline (7.5 ml/kg) i.v., and heart ventricles were examined at 72 hours. Morphometric analysis by electron microscopy revealed extensive injuries in cytosol, mitochondria, and nuclei of DOX-treated mice in both genotypes. Significantly more severely injured nuclei were observed in Mrp1(-/-) versus WT mice (P = 0.031). GSH and the GSH/GSSG ratio were significantly increased in treatment-naïve Mrp1(-/-) versus WT mice; GSH remained significantly higher in Mrp1(-/-) versus WT mice after saline and DOX treatment, with no changes in GSSG or GSH/GSSG. GS-HNE, measured by mass spectrometry, was lower in the hearts of treatment-naïve Mrp1(-/-) versus WT mice (P < 0.05). DOX treatment decreased GS-HNE in WT but not Mrp1(-/-) mice, so that GS-HNE was modestly but significantly higher in Mrp1(-/-) versus WT hearts after DOX. Expression of enzymes mediating GSH synthesis and antioxidant proteins did not differ between genotypes. Thus, despite elevated GSH levels in Mrp1(-/-) hearts, DOX induced significantly more injury in the nuclei of Mrp1(-/-) versus WT hearts.
Collapse
Affiliation(s)
- Jun Deng
- Department of Toxicology and Cancer Biology (J.D., D.C., L.C., W.Z., D.St.C., M.V.), Division of Cardiovascular Medicine (M.S., A.J.M.), Markey Cancer Center (C.W.) University of Kentucky, Lexington, Kentucky; Department of Pathology and Laboratory Medicine, William S. Middleton Memorial Veterans Administration Hospital and University of Wisconsin Medical School, Madison Wisconsin (L.C.); and School of Pharmacy, Fairleigh Dickinson University, Florham Park, New Jersey (P.J.)
| | - Donna Coy
- Department of Toxicology and Cancer Biology (J.D., D.C., L.C., W.Z., D.St.C., M.V.), Division of Cardiovascular Medicine (M.S., A.J.M.), Markey Cancer Center (C.W.) University of Kentucky, Lexington, Kentucky; Department of Pathology and Laboratory Medicine, William S. Middleton Memorial Veterans Administration Hospital and University of Wisconsin Medical School, Madison Wisconsin (L.C.); and School of Pharmacy, Fairleigh Dickinson University, Florham Park, New Jersey (P.J.)
| | - Wei Zhang
- Department of Toxicology and Cancer Biology (J.D., D.C., L.C., W.Z., D.St.C., M.V.), Division of Cardiovascular Medicine (M.S., A.J.M.), Markey Cancer Center (C.W.) University of Kentucky, Lexington, Kentucky; Department of Pathology and Laboratory Medicine, William S. Middleton Memorial Veterans Administration Hospital and University of Wisconsin Medical School, Madison Wisconsin (L.C.); and School of Pharmacy, Fairleigh Dickinson University, Florham Park, New Jersey (P.J.)
| | - Manjula Sunkara
- Department of Toxicology and Cancer Biology (J.D., D.C., L.C., W.Z., D.St.C., M.V.), Division of Cardiovascular Medicine (M.S., A.J.M.), Markey Cancer Center (C.W.) University of Kentucky, Lexington, Kentucky; Department of Pathology and Laboratory Medicine, William S. Middleton Memorial Veterans Administration Hospital and University of Wisconsin Medical School, Madison Wisconsin (L.C.); and School of Pharmacy, Fairleigh Dickinson University, Florham Park, New Jersey (P.J.)
| | - Andrew J Morris
- Department of Toxicology and Cancer Biology (J.D., D.C., L.C., W.Z., D.St.C., M.V.), Division of Cardiovascular Medicine (M.S., A.J.M.), Markey Cancer Center (C.W.) University of Kentucky, Lexington, Kentucky; Department of Pathology and Laboratory Medicine, William S. Middleton Memorial Veterans Administration Hospital and University of Wisconsin Medical School, Madison Wisconsin (L.C.); and School of Pharmacy, Fairleigh Dickinson University, Florham Park, New Jersey (P.J.)
| | - Chi Wang
- Department of Toxicology and Cancer Biology (J.D., D.C., L.C., W.Z., D.St.C., M.V.), Division of Cardiovascular Medicine (M.S., A.J.M.), Markey Cancer Center (C.W.) University of Kentucky, Lexington, Kentucky; Department of Pathology and Laboratory Medicine, William S. Middleton Memorial Veterans Administration Hospital and University of Wisconsin Medical School, Madison Wisconsin (L.C.); and School of Pharmacy, Fairleigh Dickinson University, Florham Park, New Jersey (P.J.)
| | - Luksana Chaiswing
- Department of Toxicology and Cancer Biology (J.D., D.C., L.C., W.Z., D.St.C., M.V.), Division of Cardiovascular Medicine (M.S., A.J.M.), Markey Cancer Center (C.W.) University of Kentucky, Lexington, Kentucky; Department of Pathology and Laboratory Medicine, William S. Middleton Memorial Veterans Administration Hospital and University of Wisconsin Medical School, Madison Wisconsin (L.C.); and School of Pharmacy, Fairleigh Dickinson University, Florham Park, New Jersey (P.J.)
| | - Daret St Clair
- Department of Toxicology and Cancer Biology (J.D., D.C., L.C., W.Z., D.St.C., M.V.), Division of Cardiovascular Medicine (M.S., A.J.M.), Markey Cancer Center (C.W.) University of Kentucky, Lexington, Kentucky; Department of Pathology and Laboratory Medicine, William S. Middleton Memorial Veterans Administration Hospital and University of Wisconsin Medical School, Madison Wisconsin (L.C.); and School of Pharmacy, Fairleigh Dickinson University, Florham Park, New Jersey (P.J.)
| | - Mary Vore
- Department of Toxicology and Cancer Biology (J.D., D.C., L.C., W.Z., D.St.C., M.V.), Division of Cardiovascular Medicine (M.S., A.J.M.), Markey Cancer Center (C.W.) University of Kentucky, Lexington, Kentucky; Department of Pathology and Laboratory Medicine, William S. Middleton Memorial Veterans Administration Hospital and University of Wisconsin Medical School, Madison Wisconsin (L.C.); and School of Pharmacy, Fairleigh Dickinson University, Florham Park, New Jersey (P.J.)
| | - Paiboon Jungsuwadee
- Department of Toxicology and Cancer Biology (J.D., D.C., L.C., W.Z., D.St.C., M.V.), Division of Cardiovascular Medicine (M.S., A.J.M.), Markey Cancer Center (C.W.) University of Kentucky, Lexington, Kentucky; Department of Pathology and Laboratory Medicine, William S. Middleton Memorial Veterans Administration Hospital and University of Wisconsin Medical School, Madison Wisconsin (L.C.); and School of Pharmacy, Fairleigh Dickinson University, Florham Park, New Jersey (P.J.)
| |
Collapse
|
38
|
Dorokhina EI, Magomedova AU, Shevelev AA, Kulikov SM, Gitis MK, Vedernikov AV, Vorobyev AI, Kravchenko SK. [Late cardiotoxicity of high-dose chemotherapy according to the modified NHL-BFM-90 program in adult patients with diffuse large B-cell lymphoma]. TERAPEVT ARKH 2015; 87:51-57. [PMID: 26390725 DOI: 10.17116/terarkh201587751-57] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
AIM To evaluate the late cardiotoxicity (CT) of high-dose chemotherapy (CT) according to the modified NHL-BFM-90 (mNHL-BFM-90) protocol in adult patients with diffuse large B-cell lymphoma (DLBCL). SUBJECTS AND METHODS The results of electrocardiography (ECG) and echocardiography (echoCG) were analyzed in 40 DLBCL patients treated according to the mNHL-BFM-90 program in the Hematology Research Center (HRC), Russian Academy of Medical Sciences (RAMS), in 2002 to 2009. A study group consisted of 20 men and 20 women whose age was 31 to 76 years; median age was 56.5 years at the time of their examination and the median follow-up time after therapy was 6 years. The individual cumulative dose of doxorubicin was 150-300 mg/M2. A comparison group included 19 patients receiving CHOP/R-CHOP CT in HRC, RAMS, in 2002 to 2009. Out of them, there were 8 men and 11 women whose age was 39 to 78 years median age was 70 years at the time of their examination. The individual cumulative dose of doxorubicin was 200-400 mg/M2. ECG and echoCG were carried out before and 5 years or more after CT. RESULTS Out of the 40 patients with DLBCL, the signs of subclinical cardiomyopathy (CMP) were detected in 24 (60%) patients; no clinical manifestations of congestive heart failure (CHF) were found in any patient. In the comparison group of 19 patients receiving CHOP/R-CHOP CT, 14 (74%) patients were found to have signs of subclinical CMP and no clinical signs of CHF. The summary toxicity index significantly depended on age (p=0.03) and a history of heart disease (p=0.3); it was significantly higher after CHOP/R-CHOP CT (p=0.05). There was a statistically significant relationship of the risk of subclinical CMP to the history of heart diseases (p=0.05). CONCLUSION Late cardiotoxicity of the mNHL-BFM-90 program does not exceed the toxicity of standard CHOP/R-CHOP therapy. Post-CT Echo-CG and ECG findings showed that the patients with the most marked subclinical signs of CMP in both groups had cardiotoxicity risk factors, such as coronary heart disease, hypertensive disease, or diabetes in their history. No clinically significant CHF was identified in any patient.
Collapse
Affiliation(s)
- E I Dorokhina
- Hematology Research Center, Ministry of Health of Russia, Moscow, Russia
| | - A U Magomedova
- Hematology Research Center, Ministry of Health of Russia, Moscow, Russia
| | - A A Shevelev
- Hematology Research Center, Ministry of Health of Russia, Moscow, Russia
| | - S M Kulikov
- Hematology Research Center, Ministry of Health of Russia, Moscow, Russia
| | - M K Gitis
- Hematology Research Center, Ministry of Health of Russia, Moscow, Russia
| | - A V Vedernikov
- Hematology Research Center, Ministry of Health of Russia, Moscow, Russia
| | - A I Vorobyev
- Hematology Research Center, Ministry of Health of Russia, Moscow, Russia
| | - S K Kravchenko
- Hematology Research Center, Ministry of Health of Russia, Moscow, Russia
| |
Collapse
|
39
|
Singh P, Sharma R, McElhanon K, Allen CD, Megyesi JK, Beneš H, Singh SP. Sulforaphane protects the heart from doxorubicin-induced toxicity. Free Radic Biol Med 2015; 86:90-101. [PMID: 26025579 PMCID: PMC4554811 DOI: 10.1016/j.freeradbiomed.2015.05.028] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 05/04/2015] [Accepted: 05/19/2015] [Indexed: 01/08/2023]
Abstract
Cardiotoxicity is one of the major side effects encountered during cancer chemotherapy with doxorubicin (DOX) and other anthracyclines. Previous studies have shown that oxidative stress caused by DOX is one of the primary mechanisms for its toxic effects on the heart. Since the redox-sensitive transcription factor, Nrf2, plays a major role in protecting cells from the toxic metabolites generated during oxidative stress, we examined the effects of the phytochemical sulforaphane (SFN), a potent Nrf2-activating agent, on DOX-induced cardiotoxicity. These studies were carried out both in vitro and in vivo using rat H9c2 cardiomyoblast cells and wild type 129/sv mice, and involved SFN pretreatment followed by SFN administration during DOX exposure. SFN treatment protected H9c2 cells from DOX cytotoxicity and also resulted in restored cardiac function and a significant reduction in DOX-induced cardiomyopathy and mortality in mice. Specificity of SFN induction of Nrf2 and protection of H9c2 cells was demonstrated in Nrf2 knockdown experiments. Cardiac accumulation of 4-hydroxynonenal (4-HNE) protein adducts, due to lipid peroxidation following DOX-induced oxidative stress, was significantly attenuated by SFN treatment. The respiratory function of cardiac mitochondria isolated from mice exposed to DOX alone was repressed, while SFN treatment with DOX significantly elevated mitochondrial respiratory complex activities. Co-administration of SFN reversed the DOX-associated reduction in nuclear Nrf2 binding activity and restored cardiac expression of Nrf2-regulated genes at both the RNA and protein levels. Together, our results demonstrate for the first time that the Nrf2 inducer, SFN, has the potential to provide protection against DOX-mediated cardiotoxicity.
Collapse
Affiliation(s)
- Preeti Singh
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| | - Rajendra Sharma
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Kevin McElhanon
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| | - Charles D Allen
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| | - Judit K Megyesi
- Central Arkansas Veterans Healthcare System, Little Rock, AR, USA; Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Helen Beneš
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Sharda P Singh
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; Central Arkansas Veterans Healthcare System, Little Rock, AR, USA.
| |
Collapse
|
40
|
Lin CL, Wang SE, Hsu CH, Sheu SJ, Wu CH. Oral treatment with herbal formula B307 alleviates cardiac failure in aging R6/2 mice with Huntington's disease via suppressing oxidative stress, inflammation, and apoptosis. Clin Interv Aging 2015; 10:1173-87. [PMID: 26229452 PMCID: PMC4516205 DOI: 10.2147/cia.s86493] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Cardiac failure is often observed in aging patients with Huntington’s disease (HD). However, conventional pharmacological treatments for cardiac failure in HD patients have rarely been studied. Chinese herbal medicines, especially combined herbal formulas, have been widely used to treat cardiac dysfunctions over the centuries. Thus, we assess whether oral treatment with herbal formula B307 can alleviate cardiac failure in transgenic mice with HD. After oral B307 or vehicle treatment for 2 weeks, cardiac function and cardiomyocytes in 12-week-old male R6/2 HD mice and their wild-type littermate controls (WT) were examined and then compared via echocardiography, immunohistochemistry, and Western blotting. We found that cardiac performance in aging R6/2 HD mice had significantly deteriorated in comparison with their WT (P<0.01). Cardiac expressions of superoxide dismutase 2 (SOD2) and B-cell lymphoma 2 (Bcl-2) in aging R6/2 HD mice were significantly lower than their WT (P<0.01), but cardiac expressions of tumor necrosis factor alpha (TNF-α), neurotrophin-3 (3-NT), 4-hydroxynonenal (4-HNE), Bcl-2-associated X protein (Bax), calpain, caspase 12, caspase 9, and caspase 3 of aging R6/2 HD mice were significantly higher than their WT (P<0.05). Furthermore, we found that cardiac performance in aging R6/2 HD mice had significantly improved under oral B307 treatment (P<0.05). Cardiac expressions of SOD2 and Bcl-2 of aging R6/2 HD mice were significantly higher under oral B307 treatment (P<0.01), but cardiac expressions of TNF-α, 3-NT, 4-HNE, Bax, calpain, caspase 12, caspase 9, and caspase 3 of aging R6/2 HD mice were significantly reduced under oral B307 treatment (P<0.05). Oral B307 treatment may briefly alleviate cardiac failure in aging HD R6/2 mice via suppressing cardiac oxidative stress, inflammation, and apoptosis. We suggested that the herbal formula B307 may be further developed as a potential health supplement for ameliorating cardiac failure associated with aging.
Collapse
Affiliation(s)
- Ching-Lung Lin
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Sheue-Er Wang
- Department of Pathological Inspection, Soeurs de Saint Paul de Chartres Medical Corporate Body, Taoyuan City, Taiwan
| | - Chih-Hsiang Hsu
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | | | - Chung-Hsin Wu
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| |
Collapse
|
41
|
Lien CY, Chuang TY, Hsu CH, Lin CL, Wang SE, Sheu SJ, Chien CT, Wu CH. Oral treatment with the herbal formula B307 alleviates cardiac toxicity in doxorubicin-treated mice via suppressing oxidative stress, inflammation, and apoptosis. Onco Targets Ther 2015; 8:1193-210. [PMID: 26060405 PMCID: PMC4454207 DOI: 10.2147/ott.s82936] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE This study aimed to investigate whether the herbal formula B307 could alleviate doxorubicin (DOX)-induced acute cardiotoxicity. If so, we further unraveled possible molecular mechanisms of cardiac protection under treatment with the herbal formula B307. METHODS Before the animal experiment, we examined relative viabilities of Huh7 cancer cells under treatment with the herbal formula B307. To test whether oral treatment with the herbal formula B307 could alleviate cardiotoxicity, equal volumes of B307 (50 mg/kg) or saline (sham treatment) were administered to 20-week-old male mice once daily for 14 consecutive days. Then, DOX (10 mg/kg; ip) was administered to male mice under B307 and sham treatments at 22-23 weeks of age. Cardiac functions in these mice were assessed via echocardiography at 23-24 weeks of age. Then, expressions of oxidative stress, inflammation, and apoptosis-related proteins were examined in the heart tissue by immunohistochemistry and Western blotting at 24-25 weeks of age. Apart from this, mortality rate and body weight were measured during the experiment. RESULTS In vitro, the relative viabilities of Huh7 cancer cells under treatment with the herbal formula B307 had shown no obvious change at doses of 10-160 ng/mL. Furthermore, the relative viabilities of Huh7 cancer cells were significantly reduced under DOX treatment but showed no significant change under DOX only and DOX plus B307 treatment. In vivo, the mortality rate, body weight, and cardiac function of DOX-treated mice were obviously improved under oral treatment with the herbal formula B307. Furthermore, cardiac expressions of endothelial nitric oxide synthase, superoxide dismutase 2, and B-cell lymphoma 2 were significantly enhanced, but tumor necrosis factor alpha, NFKB1 (p50 and its precursor, p105), neurotrophin-3, Bcl-2-associated X protein, calpain, caspase 12, caspase 9, and caspase 3 were significantly suppressed in DOX-treated mice under oral treatment with the herbal formula B307. CONCLUSION Our results revealed that oral treatment with the herbal formula B307 may provide cardioprotection in DOX-treated mice via suppressing oxidative stress, inflammation, and apoptosis in heart tissue. We believe that the herbal formula B307 may be developed as a potential alternative treatment for cancer patients under DOX treatment.
Collapse
Affiliation(s)
- Chia-Ying Lien
- Department of Athletics, National Taiwan University, Taipei, Taiwan
| | - Tai-Yuan Chuang
- Department of Athletics, National Taiwan University, Taipei, Taiwan
| | - Chih-Hsiang Hsu
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Ching-Lung Lin
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Sheue-Er Wang
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | | | - Chiang-Ting Chien
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Chung-Hsin Wu
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| |
Collapse
|
42
|
Chen RC, Xu XD, Zhi Liu X, Sun GB, Zhu YD, Dong X, Wang J, Zhang HJ, Zhang Q, Sun XB. Total Flavonoids from Clinopodium chinense (Benth.) O. Ktze Protect against Doxorubicin-Induced Cardiotoxicity In Vitro and In Vivo. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2015; 2015:472565. [PMID: 25784945 PMCID: PMC4346128 DOI: 10.1155/2015/472565] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Revised: 12/04/2014] [Accepted: 12/22/2014] [Indexed: 01/08/2023]
Abstract
Doxorubicin has cardiotoxic effects that limit its clinical benefit in cancer patients. This study aims to investigate the protective effects of the total flavonoids from Clinopodium chinense (Benth.) O. Ktze (TFCC) against doxorubicin- (DOX-) induced cardiotoxicity. Male rats were intraperitoneally injected with a single dose of DOX (3 mg/kg) every 2 days for three injections. Heart samples were collected 2 weeks after the last DOX dose and then analyzed. DOX delayed body and heart growth and caused cardiac tissue injury, oxidative stress, apoptotic damage, mitochondrial dysfunction, and Bcl-2 expression disturbance. Similar experiments in H9C2 cardiomyocytes showed that doxorubicin reduced cell viability, increased ROS generation and DNA fragmentation, disrupted mitochondrial membrane potential, and induced apoptotic cell death. However, TFCC pretreatment suppressed all of these adverse effects of doxorubicin. Signal transduction studies indicated that TFCC suppressed DOX-induced overexpression of p53 and phosphorylation of JNK, p38, and ERK. Studies with LY294002 (a PI3K/AKT inhibitor) demonstrated that the mechanism of TFCC-induced cardioprotection also involves activation of PI3K/AKT. These findings indicated the potential clinical application of TFCC in preventing DOX-induced cardiac oxidative stress.
Collapse
Affiliation(s)
- Rong Chang Chen
- Institute of Medicinal Plant Development, Chinese Academy of Medical Science, Peking Union Medical College, No. 151, North Road Malianwa, Haidian District, Beijing 100094, China
| | - Xu Dong Xu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Science, Peking Union Medical College, No. 151, North Road Malianwa, Haidian District, Beijing 100094, China
| | - Xue Zhi Liu
- Academy of Forestry, Baishan, Jilin 134302, China
| | - Gui Bo Sun
- Institute of Medicinal Plant Development, Chinese Academy of Medical Science, Peking Union Medical College, No. 151, North Road Malianwa, Haidian District, Beijing 100094, China
| | - Yin Di Zhu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Science, Peking Union Medical College, No. 151, North Road Malianwa, Haidian District, Beijing 100094, China
| | - Xi Dong
- Academy of Chinese Materia Medica, Wenzhou Medical College, Wenzhou, Zhejiang 325035, China
| | - Jian Wang
- Harbin University of Commerce, Xuehai Street, Songbei District, Harbin, Heilongjiang 150028, China
| | - Hai Jing Zhang
- Department of Pharmacology, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, China
| | - Qiang Zhang
- Academy of Chinese Materia Medica, Wenzhou Medical College, Wenzhou, Zhejiang 325035, China
| | - Xiao Bo Sun
- Institute of Medicinal Plant Development, Chinese Academy of Medical Science, Peking Union Medical College, No. 151, North Road Malianwa, Haidian District, Beijing 100094, China
| |
Collapse
|
43
|
Beneš H, Vuong MK, Boerma M, McElhanon KE, Siegel ER, Singh SP. Protection from oxidative and electrophilic stress in the Gsta4-null mouse heart. Cardiovasc Toxicol 2014; 13:347-56. [PMID: 23690225 DOI: 10.1007/s12012-013-9215-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
4-Hydroxynonenal (4-HNE) mediates many pathological effects of oxidative and electrophilic stress and signals to activate cytoprotective gene expression regulated by NF-E2-related factor 2 (Nrf2). By exhibiting very high levels of 4-HNE-conjugating activity, the murine glutathione transferase alpha 4 (GSTA4-4) helps regulate cellular 4-HNE levels. To examine the role of 4-HNE in vivo, we disrupted the murine Gsta4 gene. Gsta4-null mice exhibited no cardiac phenotype under normal conditions and no difference in cardiac 4-HNE level as compared to wild-type mice. We hypothesized that the Nrf2 pathway might contribute an important compensatory mechanism to remove excess cardiac 4-HNE in Gsta4-null mice. Cardiac nuclear extracts from Gsta4-null mice exhibited significantly higher Nrf2 binding to antioxidant response elements. We also observed responses in critical Nrf2 target gene products: elevated Sod2, Cat, and Akr1b7 mRNA levels and significant increases in both cardiac antioxidant and anti-electrophile enzyme activities. Gsta4-null mice were less sensitive and maintained normal cardiac function following chronic doxorubicin treatment, known to increase cardiac 4-HNE levels. Hence, in the absence of GSTA4-4 to modulate both physiological and pathological 4-HNE levels, the adaptive Nrf2 pathway may be primed to contribute to a preconditioned cardiac phenotype in the Gsta4-null mouse.
Collapse
Affiliation(s)
- Helen Beneš
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | | | | | | | | | | |
Collapse
|
44
|
Zhao Y, Miriyala S, Miao L, Mitov M, Schnell D, Dhar SK, Cai J, Klein JB, Sultana R, Butterfield DA, Vore M, Batinic-Haberle I, Bondada S, St Clair DK. Redox proteomic identification of HNE-bound mitochondrial proteins in cardiac tissues reveals a systemic effect on energy metabolism after doxorubicin treatment. Free Radic Biol Med 2014; 72:55-65. [PMID: 24632380 PMCID: PMC4053505 DOI: 10.1016/j.freeradbiomed.2014.03.001] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 02/17/2014] [Accepted: 03/04/2014] [Indexed: 10/25/2022]
Abstract
Doxorubicin (DOX), one of the most effective anticancer drugs, is known to generate progressive cardiac damage, which is due, in part, to DOX-induced reactive oxygen species (ROS). The elevated ROS often induce oxidative protein modifications that result in alteration of protein functions. This study demonstrates that the level of proteins adducted by 4-hydroxy-2-nonenal (HNE), a lipid peroxidation product, is significantly increased in mouse heart mitochondria after DOX treatment. A redox proteomics method involving two-dimensional electrophoresis followed by mass spectrometry and investigation of protein databases identified several HNE-modified mitochondrial proteins, which were verified by HNE-specific immunoprecipitation in cardiac mitochondria from the DOX-treated mice. The majority of the identified proteins are related to mitochondrial energy metabolism. These include proteins in the citric acid cycle and electron transport chain. The enzymatic activities of the HNE-adducted proteins were significantly reduced in DOX-treated mice. Consistent with the decline in the function of the HNE-adducted proteins, the respiratory function of cardiac mitochondria as determined by oxygen consumption rate was also significantly reduced after DOX treatment. Treatment with Mn(III) meso-tetrakis(N-n-butoxyethylpyridinium-2-yl)porphyrin, an SOD mimic, averted the doxorubicin-induced mitochondrial dysfunctions as well as the HNE-protein adductions. Together, the results demonstrate that free radical-mediated alteration of energy metabolism is an important mechanism mediating DOX-induced cardiac injury, suggesting that metabolic intervention may represent a novel approach to preventing cardiac injury after chemotherapy.
Collapse
Affiliation(s)
- Y Zhao
- Graduate Center for Toxicology, University of Kentucky, Lexington, KY 40506, USA
| | - S Miriyala
- Graduate Center for Toxicology, University of Kentucky, Lexington, KY 40506, USA; Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences, Shreveport, LA 71130, USA
| | - L Miao
- Graduate Center for Toxicology, University of Kentucky, Lexington, KY 40506, USA
| | - M Mitov
- Free Radical Biology in Cancer Shared Resource Facility, Markey Cancer Center, University of Kentucky, Lexington, KY 40506, USA
| | - D Schnell
- Graduate Center for Toxicology, University of Kentucky, Lexington, KY 40506, USA
| | - S K Dhar
- Graduate Center for Toxicology, University of Kentucky, Lexington, KY 40506, USA
| | - J Cai
- Department of Nephrology and Proteomics Facility, University of Louisville, Louisville, KY 40292, USA
| | - J B Klein
- Department of Nephrology and Proteomics Facility, University of Louisville, Louisville, KY 40292, USA
| | - R Sultana
- Department of Chemistry, Center of Membrane Sciences, and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA
| | - D A Butterfield
- Free Radical Biology in Cancer Shared Resource Facility, Markey Cancer Center, University of Kentucky, Lexington, KY 40506, USA; Department of Chemistry, Center of Membrane Sciences, and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA
| | - M Vore
- Graduate Center for Toxicology, University of Kentucky, Lexington, KY 40506, USA
| | - I Batinic-Haberle
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA
| | - S Bondada
- Department of Immunology, University of Kentucky, Lexington, KY 40506, USA
| | - D K St Clair
- Graduate Center for Toxicology, University of Kentucky, Lexington, KY 40506, USA.
| |
Collapse
|
45
|
Substitution of soy protein for casein prevents oxidative modification and inflammatory response induced in rats fed high fructose diet. ISRN INFLAMMATION 2014; 2014:641096. [PMID: 25006525 PMCID: PMC4009313 DOI: 10.1155/2014/641096] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 03/16/2014] [Indexed: 11/18/2022]
Abstract
Fructose-rich diet is known to cause metabolic dysregulation, oxidative stress, and inflammation. We aimed to compare the effects of two dietary proteins of animal and plant origins on fructose-induced oxidative stress and inflammatory changes in liver. Wistar rats were fed either starch or fructose (60%) diet with casein or soy protein (20%) as the protein source for 8 weeks. Glucose and insulin, glycated hemoglobin and fructosamine, AOPP, and FRAP were determined in circulation. Intracellular ROS, oxidatively modified proteins (4-HNE and 3-NT adducts), adiponectin, TNF-α, IL-6 and PAI-1 mRNA expression, phosphorylation and activation of JNK and IKKβ, and NF-κB binding activity were assayed in liver. In comparison with starch fed group, fructose + casein group registered significant decline in antioxidant potential and increase in plasma glucose, insulin, and glycated proteins. Increased ROS production, 4-HNE and 3-NT modified proteins, JNK and IKKβ activation, and NF-κB binding activity were observed in them along with increased gene expression of PAI-1, IL-6, and TNF-α and decreased adiponectin expression. Substitution of soy protein for casein reduced oxidative modification and inflammatory changes in fructose-fed rats. These data suggest that soy protein but not casein can avert the adverse effects elicited by chronic consumption of fructose.
Collapse
|
46
|
Cole MP, Tangpong J, Oberley TD, Chaiswing L, Kiningham KK, St. Clair DK. Nuclear interaction between ADR-induced p65 and p53 mediates cardiac injury in iNOS (-/-) mice. PLoS One 2014; 9:e89251. [PMID: 24586632 PMCID: PMC3934890 DOI: 10.1371/journal.pone.0089251] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 01/17/2014] [Indexed: 11/20/2022] Open
Abstract
Adriamycin (ADR) treatment causes an imbalance in the levels of nitric oxide (•NO) and superoxide (O2•−) production leading to cardiac injury. Previously we demonstrated that mice lacking inducible nitric oxide synthase (iNOS) have increased oxidative stress and mitochondrial injury. The molecular events leading to increased mitochondrial injury in iNOS deficient mice is unknown. ADR in the absence of iNOS preferentially activates a proapoptotic pathway without a concurrent increase in prosurvival pathways. Treatment with ADR leads to an increase in DNA binding activity of nuclear factor kappa B (NFκB) and p53 in wildtype mice. Following ADR treatment, p53, but not NFκB DNA binding activity, as well as the level of Bax, a p53 target gene, was increased in iNOS (−/−) mice. This apoptotic signaling effect in iNOS (−/−) is alleviated by overexpression of manganese superoxide dismutase (MnSOD). Increases in NFκB and p53 in ADR-treated wildtype mice did not lead to increases in target genes such as MnSOD, bcl-xL, or Bax. Moreover, co-immunoprecipitation analysis revealed that p65, a prominent member of the NFκB family, interacts with p53 in the nucleus. These results suggest that NFκB and p53 may counter act one another's actions in ADR-treated wildtype (WT) mice. Further, these results identify a novel mechanism by which oxidative stress may regulate transcription of proapoptotic genes.
Collapse
Affiliation(s)
- Marsha P. Cole
- Biochemistry and Molecular Biology, University of Louisville, Louisville, Kentucky, United States of America
- * E-mail:
| | - Jitbanjong Tangpong
- School of Allied Health Sciences and Public Health, Walailak University, Nakhon Si Thammarat, Thailand
| | - Terry D. Oberley
- Department of Pathology, VA Hospital, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Luksana Chaiswing
- Department of Pathology, VA Hospital, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Kinsley K. Kiningham
- Pharmaceutical, Social and Administrative Sciences, Belmont College of Pharmacy, Nashville, Tennessee, United States of America
| | - Daret K. St. Clair
- Graduate Centers for Toxicology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, United States of America
| |
Collapse
|
47
|
Oktem G, Uysal A, Oral O, Sezer ED, Olukman M, Erol A, Akgur SA, Bilir A. Resveratrol attenuates doxorubicin-induced cellular damage by modulating nitric oxide and apoptosis. ACTA ACUST UNITED AC 2012; 64:471-9. [DOI: 10.1016/j.etp.2010.11.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2009] [Revised: 06/22/2010] [Accepted: 11/01/2010] [Indexed: 01/19/2023]
|
48
|
He L, Xiao J, Fu H, Du G, Xiao X, Zhang C, Gu Y, Ma Y. Effect of oxidative stress on ventricular arrhythmia in rabbits with adriamycin-induced cardiomyopathy. ACTA ACUST UNITED AC 2012; 32:334-339. [PMID: 22684554 DOI: 10.1007/s11596-012-0058-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2011] [Indexed: 01/01/2023]
Abstract
The purpose of the present study was to examine the effects of oxidative stress on ventricular arrhythmias in rabbits with adriamycin-induced cardiomyopathy and the relationship between oxidative stress and ventricular arrhythmia. Forty Japanese white rabbits were randomly divided into four groups (n=10 in each): control group, metoprolol (a selective β1 receptor blocker) group, carvedilol (a nonselective β blocker/α-1 blocker) group and adriamycin group. Models of adriamycin-induced cardiomyopathy were established by intravenously injecting adriamycin hydrochloride (1 mg/kg) to rabbits via the auri-edge vein twice a week for 8 weeks in the adriamycin, metoprolol and carvedilol groups. Rabbits in the control group were given equal volume of saline through the auri-edge vein. Rabbits in the metoprolol and carvedilol groups were then intragastrically administrated metoprolol (5 mg/kg/d) and carvedilol (5 mg/kg/d) respectively for 2 months, while those in the adriamycin and control groups were treated with equal volume of saline in the same manner as in the metroprolol and carvedilol groups. Left ventricular end diastolic diameter (LVEDd) and left ventricular ejection fraction (LVEF) were measured by echocardiography. Plasma levels of N-terminal pro B-type natriuretic peptide (NT-proBNP), malondialdehyde (MAD) and superoxide dismutase (SOD) were detected. The left ventricular wedge preparations were perfused with Tyrode's solution. The transmural electrocardiogram, transmural action potentials from epicardium (Epi) and endocardium (Endo), transmural repolarization dispersion (TDR) were recorded, and the incidences of triggered activity and ventricular arrhythmias were obtained at rapid cycle lengths. The results showed that TDR and the serum MDA and NT-proBNP levels were increased, and LVEF and the serum SOD level decreased in the adriamycin group compared with the control group. The incidences of triggered activity and ventricular arrhythmia were significantly higher in the adriamycin group than those in the control group (P<0.05). In the carvedilol group as compared with the adriamycin group, the serum SOD level and the LVEF were substantially increased; the TDR, and the serum MDA and NT-proBNP levels were significantly decreased; the incidences of triggered activity and ventricular arrhythmia were obviously reduced (P<0.05). There were no significant differences in the levels of MDA and SOD, LVEF, TDR and the incidences of triggered activity and ventricular arrhythmia between the adriamycin group and the metoprolol group. It was concluded that carvedilol may inhibit triggered activity and ventricular arrhythmias in rabbit with adriamycin-induced cardiomyopathy, which is related to the decrease in oxygen free radials.
Collapse
Affiliation(s)
- Li He
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Department of Cardiology, Puai Hospital, Wuhan, 430033, China
| | - Jianmin Xiao
- Department of Cardiology, Dongguan Taiping Hospital, Dongguan, 523905, China
| | - Hui Fu
- Department of Pharmacology, Guangdong Medical College, Guangzhou, 523808, China
| | - Guangsheng Du
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xing Xiao
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Cuntai Zhang
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ye Gu
- Department of Cardiology, Puai Hospital, Wuhan, 430033, China
| | - Yexin Ma
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
49
|
Comparative study on the effects of grape seed extract and telmisartan on doxorubicin-induced cardiotoxicity in adult male rats. ACTA ACUST UNITED AC 2012. [DOI: 10.1097/01.ehx.0000414803.54664.e5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
50
|
Holley AK, Dhar SK, Xu Y, St. Clair DK. Manganese superoxide dismutase: beyond life and death. Amino Acids 2012; 42:139-58. [PMID: 20454814 PMCID: PMC2975048 DOI: 10.1007/s00726-010-0600-9] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2009] [Accepted: 04/16/2010] [Indexed: 02/07/2023]
Abstract
Manganese superoxide dismutase (MnSOD) is a nuclear-encoded antioxidant enzyme that localizes to the mitochondria. Expression of MnSOD is essential for the survival of aerobic life. Transgenic mice expressing a luciferase reporter gene under the control of the human MnSOD promoter demonstrate that the level of MnSOD is reduced prior to the formation of cancer. Overexpression of MnSOD in transgenic mice reduces the incidences and multiplicity of papillomas in a DMBA/TPA skin carcinogenesis model. However, MnSOD deficiency does not lead to enhanced tumorigenicity of skin tissue similarly treated because MnSOD can modulate both the p53-mediated apoptosis and AP-1-mediated cell proliferation pathways. Apoptosis is associated with an increase in mitochondrial levels of p53 suggesting a link between MnSOD deficiency and mitochondrial-mediated apoptosis. Activation of p53 is preventable by application of a SOD mimetic (MnTE-2-PyP(5+)). Thus, p53 translocation to mitochondria and subsequent inactivation of MnSOD explain the observed mitochondrial dysfunction that leads to transcription-dependent mechanisms of p53-induced apoptosis. Administration of MnTE-2-PyP(5+) following apoptosis but prior to proliferation leads to suppression of protein carbonyls and reduces the activity of AP-1 and the level of the proliferating cellular nuclear antigen, without reducing the activity of p53 or DNA fragmentation following TPA treatment. Remarkably, the incidence and multiplicity of skin tumors are drastically reduced in mice that receive MnTE-2-PyP(5+) prior to cell proliferation. The results demonstrate the role of MnSOD beyond its essential role for survival and suggest a novel strategy for an antioxidant approach to cancer intervention.
Collapse
Affiliation(s)
| | | | - Yong Xu
- University of Kentucky, Lexington, USA
| | | |
Collapse
|