1
|
Mahadik SR, Reddy ART, Choudhary K, Nama L, Jamdade MS, Singh S, Murti K, Kumar N. Arsenic induced cardiotoxicity: An approach for molecular markers, epigenetic predictors and targets. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2024; 111:104558. [PMID: 39245244 DOI: 10.1016/j.etap.2024.104558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 08/15/2024] [Accepted: 08/31/2024] [Indexed: 09/10/2024]
Abstract
Arsenic, a ubiquitous environmental toxicant, has been acknowledged as a significant issue for public health due to its widespread pollution of drinking water and food supplies. The present review aimed to study the toxicity associated with the cardiac system. Prolonged exposure to arsenic has been associated with several harmful health outcomes, especially cardiotoxicity. Arsenic-induced cardiotoxicity encompasses a range of cardiovascular abnormalities, including cardiac arrhythmias, ischemic heart disease, and cardiomyopathy. To tackle this toxicity, understanding the molecular markers, epigenetic predictors, and targets involved in arsenic-induced cardiotoxicity is essential for creating preventative and therapeutic approaches. For preventive measures against this heavy metal poisoning of groundwater, it is crucial to regularly monitor water quality, re-evaluate scientific findings, and educate the public about the possible risks. This review thoroughly summarised what is currently known in this field, highlighting the key molecular markers, epigenetic modifications, and potential therapeutic targets associated with arsenic-induced cardiotoxicity.
Collapse
Affiliation(s)
- Sakshi Ramesh Mahadik
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, Vaishali, Bihar 844102, India
| | - Annem Ravi Teja Reddy
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, Vaishali, Bihar 844102, India
| | - Khushboo Choudhary
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, Vaishali, Bihar 844102, India
| | - Lokesh Nama
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, Vaishali, Bihar 844102, India
| | - Mohini Santosh Jamdade
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, Vaishali, Bihar 844102, India
| | - Sanjiv Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, Vaishali, Bihar 844102, India
| | - Krishna Murti
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research, Hajipur, Vaishali, Bihar 844102, India
| | - Nitesh Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, Vaishali, Bihar 844102, India.
| |
Collapse
|
2
|
Yan M, Wang H, Wei R, Li W. Arsenic trioxide: applications, mechanisms of action, toxicity and rescue strategies to date. Arch Pharm Res 2024; 47:249-271. [PMID: 38147202 DOI: 10.1007/s12272-023-01481-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 12/15/2023] [Indexed: 12/27/2023]
Abstract
Arsenical medicine has obtained its status in traditional Chinese medicine for more than 2,000 years. In the 1970s, arsenic trioxide was identified to have high efficacy and potency for the treatment of acute promyelocytic leukemia, which promoted many studies on the therapeutic effects of arsenic trioxide. Currently, arsenic trioxide is widely used to treat acute promyelocytic leukemia and various solid tumors through various mechanisms of action in clinical practice; however, it is accompanied by a series of adverse reactions, especially cardiac toxicity. This review presents a comprehensive overview of arsenic trioxide from preclinical and clinical efficacy, potential mechanisms of action, toxicities, and rescue strategies for toxicities to provide guidance or assistance for the clinical application of arsenic trioxide.
Collapse
Affiliation(s)
- Meng Yan
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China.
| | - Hao Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Rui Wei
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
- Pharmacy Department, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Wenwen Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
3
|
Wang J, Liu YM, Hu J, Chen C. Potential of natural products in combination with arsenic trioxide: Investigating cardioprotective effects and mechanisms. Biomed Pharmacother 2023; 162:114464. [PMID: 37060657 DOI: 10.1016/j.biopha.2023.114464] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/25/2023] [Accepted: 02/28/2023] [Indexed: 04/17/2023] Open
Abstract
Over the past few decades, clinical trials conducted worldwide have demonstrated the efficacy of arsenic trioxide (ATO) in the treatment of relapsed acute promyelocytic leukemia (APL). Currently, ATO has become the frontline treatments for patients with APL. However, its therapeutic applicability is severely constrained by ATO-induced cardiac side effects. Any cardioprotective agents that can ameliorate the cardiac side effects and allow exploiting the full therapeutic potential of ATO, undoubtedly gain significant attention. The knowledge and use of natural products for evidence-based therapy have grown rapidly in recent years. Here we discussed the potential mechanism of ATO-induced cardiac side effects and reviewed the studies on cardiac side effects as well as the research history of ATO in the treatment of APL. Then, We summarized the protective effects and underlying mechanisms of natural products in the treatment of ATO-induced cardiac side effects. Based on the efficacy and safety of the natural product, it has a promising future in the development of cardioprotective agents against ATO-induced cardiac side effects.
Collapse
Affiliation(s)
- Jie Wang
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| | - Yong-Mei Liu
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| | - Jun Hu
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China.
| | - Cong Chen
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China.
| |
Collapse
|
4
|
Ahmad S, Tan M, Hamid S. DNA repair mechanisms: Exploring potentials of nutraceutical. J Funct Foods 2023. [DOI: 10.1016/j.jff.2023.105415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
|
5
|
Li Y, Wan R, Liu J, Liu W, Ma L, Zhang H. In silico mechanisms of arsenic trioxide-induced cardiotoxicity. Front Physiol 2022; 13:1004605. [PMID: 36589437 PMCID: PMC9798418 DOI: 10.3389/fphys.2022.1004605] [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: 07/27/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022] Open
Abstract
It has been found that arsenic trioxide (ATO) is effective in treating acute promyelocytic leukemia (APL). However, long QT syndrome was reported in patients receiving therapy using ATO, which even led to sudden cardiac death. The underlying mechanisms of ATO-induced cardiotoxicity have been investigated in some biological experiments, showing that ATO affects human ether-à-go-go-related gene (hERG) channels, coding rapid delayed rectifier potassium current (I Kr ), as well as L-type calcium (I CaL ) channels. Nevertheless, the mechanism by which these channel reconstitutions induced the arrhythmia in ventricular tissue remains unsolved. In this study, a mathematical model was developed to simulate the effect of ATO on ventricular electrical excitation at cellular and tissue levels by considering ATO's effects on I Kr and I CaL . The ATO-dose-dependent pore block model was incorporated into the I Kr model, and the enhanced degree of ATO to I CaL was based on experimental data. Simulation results indicated that ATO extended the action potential duration of three types of ventricular myocytes (VMs), including endocardial cells (ENDO), midmyocardial cells (MCELL), and epicardial cells (EPI), and exacerbated the heterogeneity among them. ATO could also induce alternans in all three kinds of VMs. In a cable model of the intramural ventricular strand, the effects of ATO are reflected in a prolonged QT interval of simulated pseudo-ECG and a wide vulnerable window, thus increasing the possibility of spiral wave formation in ventricular tissue. In addition to showing that ATO prolonged QT, we revealed that the heterogeneity caused by ATO is also an essential hazard factor. Based on this, a pharmacological intervention of ATO toxicity by resveratrol was undertaken. This study provides a further understanding of ATO-induced cardiotoxicity, which may help to improve the treatment for APL patients.
Collapse
Affiliation(s)
- Yacong Li
- Beijing Academy of Artificial Intelligence, Beijing, China
| | - Runlan Wan
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jun Liu
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin, China,Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China,*Correspondence: Jun Liu, ; Weichao Liu, ; Henggui Zhang,
| | - Weichao Liu
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China,*Correspondence: Jun Liu, ; Weichao Liu, ; Henggui Zhang,
| | - Lei Ma
- Beijing Academy of Artificial Intelligence, Beijing, China,National Biomedical Imaging Center, Peking University, Beijing, China
| | - Henggui Zhang
- Biological Physics Group, School of Physics and Astronomy, The University of Manchester, Manchester, United Kingdom,*Correspondence: Jun Liu, ; Weichao Liu, ; Henggui Zhang,
| |
Collapse
|
6
|
Orta Yilmaz B, Aydin Y. Disruption of Leydig cell steroidogenic function by sodium arsenite and/or sodium fluoride. Theriogenology 2022; 193:146-156. [PMID: 36182826 DOI: 10.1016/j.theriogenology.2022.09.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 09/08/2022] [Accepted: 09/19/2022] [Indexed: 11/18/2022]
Abstract
Arsenite (As) and fluoride (F), both of which are linked to a variety of human ailments, are regularly found in underground drinking water. Numerous studies have shown that As and/or F have negative impacts on testicular function and fertility. For this purpose, mouse Leydig cells, the main cells responsible for the generation and regulation of steroid hormones such as testosterone, were used to reveal the effects of individual and combined exposure of As and F on the steroidogenic pathway in the male reproductive system. Leydig cells were treated with 0.39 μM (50 ppb) As and 0.0476 mM (2 ppm) F alone and in combination for 24 h. The findings revealed that As and/or F exposure induced oxidative stress and apoptosis in Leydig cells and altered antioxidant equilibrium of the cells by reducing superoxide dismutase, catalase, glutathione peroxidase. Additionally, individual and combined administration of As and/or F significantly supressed the expression of both steroidogenic enzymes and the genes encoding these enzymes. In conclusion, this study showed that exposure to As and F at environmentally relevant concentrations dispersed by water decreased testosterone production in Leydig cells, an important cell of the male reproductive system. The deleterious effects of even the lowest concentrations of As and F elements that can reach humans from the environment on the Leydig cell, and therefore on male infertility, emphasize necessity new safe limits for these elements.
Collapse
Affiliation(s)
- Banu Orta Yilmaz
- Department of Biology, Faculty of Science, Istanbul University, Istanbul, Turkey.
| | - Yasemin Aydin
- Department of Biology, Faculty of Science, Istanbul University, Istanbul, Turkey
| |
Collapse
|
7
|
Adhikari A, Asdaq SMB, Al Hawaj MA, Chakraborty M, Thapa G, Bhuyan NR, Imran M, Alshammari MK, Alshehri MM, Harshan AA, Alanazi A, Alhazmi BD, Sreeharsha N. Anticancer Drug-Induced Cardiotoxicity: Insights and Pharmacogenetics. Pharmaceuticals (Basel) 2021; 14:ph14100970. [PMID: 34681194 PMCID: PMC8539940 DOI: 10.3390/ph14100970] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 09/09/2021] [Accepted: 09/21/2021] [Indexed: 12/29/2022] Open
Abstract
The advancement in therapy has provided a dramatic improvement in the rate of recovery among cancer patients. However, this improved survival is also associated with enhanced risks for cardiovascular manifestations, including hypertension, arrhythmias, and heart failure. The cardiotoxicity induced by chemotherapy is a life-threatening consequence that restricts the use of several chemotherapy drugs in clinical practice. This article addresses the prevalence of cardiotoxicity mediated by commonly used chemotherapeutic and immunotherapeutic agents. The role of susceptible genes and radiation therapy in the occurrence of cardiotoxicity is also reviewed. This review also emphasizes the protective role of antioxidants and future perspectives in anticancer drug-induced cardiotoxicities.
Collapse
Affiliation(s)
- Archana Adhikari
- Pharmacology Department, Himalayan Pharmacy Institute Majhitar, Rangpo 737136, Sikkim, India; (A.A.); (G.T.)
| | - Syed Mohammed Basheeruddin Asdaq
- Department of Pharmacy Practice, College of Pharmacy, AlMaarefa University, Dariyah, Riyadh 13713, Saudi Arabia
- Correspondence: (S.M.B.A.); (M.C.)
| | - Maitham A. Al Hawaj
- Department of Pharmacy Practice, College of Clinical Pharmacy, King Faisal University, Hofuf 31982, Saudi Arabia;
| | - Manodeep Chakraborty
- Pharmacology Department, Himalayan Pharmacy Institute Majhitar, Rangpo 737136, Sikkim, India; (A.A.); (G.T.)
- Correspondence: (S.M.B.A.); (M.C.)
| | - Gayatri Thapa
- Pharmacology Department, Himalayan Pharmacy Institute Majhitar, Rangpo 737136, Sikkim, India; (A.A.); (G.T.)
| | - Nihar Ranjan Bhuyan
- Department of Pharmaceutical Analysis, Himalayan Pharmacy Institute, Majhitar, Rangpo 737136, Sikkim, India;
| | - Mohd. Imran
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia;
| | | | - Mohammed M. Alshehri
- Pharmaceutical Care Department, Ministry of National Guard-Health Affairs, Riyadh 11426, Saudi Arabia;
| | - Aishah Ali Harshan
- Department of Pharmaceutical Care, Northern Area Armed Forces Hospital, King Khalid Military City Hospital, Hafr Al-Batin 39745, Saudi Arabia;
| | - Abeer Alanazi
- Department of Pharmaceutical Care, First Health Cluster in Eastern Province, King Fahad Specialist Hospital, Dammam 32253, Saudi Arabia;
| | | | - Nagaraja Sreeharsha
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa-31982, Saudi Arabia;
- Department of Pharmaceutics, Vidya Siri College of Pharmacy, Off Sarjapura Road, Bengaluru 560035, Karnataka, India
| |
Collapse
|
8
|
Binu P, Soman R, Zakhariah Hisham O, Narayanan SP, Nair RH. Acute promyelocytic leukemia drug - arsenic trioxide in the presence of eugenol shows differential action on leukemia cells (HL-60) and cardiomyocytes (H9c2) - inference from NMR study. Toxicol Mech Methods 2021; 31:457-466. [PMID: 33879037 DOI: 10.1080/15376516.2021.1913685] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The increased concern of cardiovascular dysfunction by cancer therapeutics has led to more effective treatment strategies. Arsenic trioxide (As2O3) is a potential chemotherapeutic agent for acute promyelocytic leukemia (APL), but the effectiveness is affected by potential cardiotoxicity. Researchers have been trying to find out novel modalities to manage the adverse effects of As2O3. In our study, the antioxidant molecule eugenol showed protective action against the destructive impact of As2O3 on cardiomyocytes (H9c2) without compromising the anti-cancer property As2O3 on leukemia cells (HL-60). We have studied the interaction between arsenic and eugenol in physiological and acidic pH to understand the molecular mechanism of differential action of As2O3 in the presence of eugenol using NMR spectroscopy. The study observed that at physiological pH, arsenic and eugenol interact to form an inactive product, positively affecting H9c2 cardiomyocytes. Still, there is no such interaction in acidic pH evidenced by the useful anti-cancer property of As2O3. The result concludes that the antioxidant molecule eugenol is an efficient protective agent against the adverse effect of As2O3 on cardiomyocytes.
Collapse
Affiliation(s)
- Prakash Binu
- Physiology Research Laboratory, School of Biosciences, Mahatma Gandhi University, Kottayam, Kerala, India
| | - Reshma Soman
- Physiology Research Laboratory, School of Biosciences, Mahatma Gandhi University, Kottayam, Kerala, India
| | - Omar Zakhariah Hisham
- NMR Facility, Institute for Integrated Programmes and Research in Basic Sciences, Mahatma Gandhi University, Kottayam, Kerala, India
| | | | | |
Collapse
|
9
|
Zhao Z, Li J, Zheng B, Liang Y, Shi J, Zhang J, Han X, Chu L, Chu X, Gao Y. Ameliorative effects and mechanism of crocetin in arsenic trioxide‑induced cardiotoxicity in rats. Mol Med Rep 2020; 22:5271-5281. [PMID: 33173984 PMCID: PMC7646993 DOI: 10.3892/mmr.2020.11587] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 08/20/2020] [Indexed: 01/15/2023] Open
Abstract
Arsenic trioxide (ATO) is commonly used to treat patients with acute promyelocytic leukemia since it was authorized by the U.S. Food and Drug Administration in the 1970s, but its applicability has been limited by its cardiotoxic effects. Therefore, the aim of the present study was to investigate the cardioprotective effects and underlying mechanism of crocetin (CRT), the critical ingredient of saffron. Sprague-Dawley rats were then randomly divided into four groups (n=10/group): i) Control group; ii) ATO group, iii) CRT-low (20 mg/kg) group; and iv) CRT-high (40 mg/kg) group. Rats in the Control and ATO groups were intraperitoneally injected with equal volumes of 0.9% sodium chloride solution, and CRT groups were administered with either 20 and 40 mg/kg CRT. Following 6 h, all groups except the Control group were intraperitoneally injected with 5 mg/kg ATO over 10 days. Cardiotoxicity was indicated by changes in electrocardiographic (ECG) patterns, morphology and marker enzymes. Histomorphological changes in the heart tissue were observed by pathological staining. The levels of superoxide dismutase, glutathione peroxidase, malondialdehyde and catalase in the serum were analyzed using colometric commercial assay kits, and the levels of reactive oxygen species in the heart tissue were detected using the fluorescent probe dihydroethidium. The expression levels of inflammatory factors and activities of apoptosis-related proteins were analyzed using immunohistochemistry. The protein expression levels of silent information regulator of transcription 1 were measured using western blotting. Cardiotoxicity was induced in male Sprague-Dawley rats with ATO (5 mg/kg). CRT (20 and 40 mg/kg) and ATO were co-administered to evaluate possible cardioprotective effects. CRT significantly reduced the heart rate and J-point elevation induced by ATO in rats. Histological changes were evaluated via hematoxylin and eosin staining. CRT decreased the levels of creatine kinase and lactate dehydrogenase, increased the activities of superoxide dismutase, glutathione-peroxidase and catalase, and decreased the levels of malondialdehyde and reactive oxygen species. Moreover, CRT downregulated the expression levels of the pro-inflammatory factors IL-1, TNF-α, IL-6, Bax and p65, as well as increased the expression of Bcl-2. It was also identified that CRT enhanced silent information regulator of transcription 1 protein expression. Thus, the present study demonstrated that CRT treatment effectively ameliorated ATO-induced cardiotoxicity. The protective effects of CRT can be attributed to the inhibition of oxidative stress, inflammation and apoptosis. Therefore, CRT represents a promising therapeutic method for improving the cardiotoxic side effects caused by ATO treatment, and additional clinical applications are possible, but warrant further investigation.
Collapse
Affiliation(s)
- Zhifeng Zhao
- Department of Pharmaceutics, School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Jinghan Li
- Department of Preventive Medicine, School of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Bin Zheng
- Department of Pharmaceutics, School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Yingran Liang
- Department of Pharmaceutics, School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Jing Shi
- Department of Pharmacy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Jianping Zhang
- Hebei Key Laboratory of Integrative Medicine on Liver‑Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Xue Han
- Department of Pharmaceutics, School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Li Chu
- Department of Pharmaceutics, School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Xi Chu
- Department of Pharmacy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Yonggang Gao
- Department of Preventive Medicine, School of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| |
Collapse
|
10
|
Diab KA, Ibrahim NE, Fahmy MA, Hassan EM, Omara EA. Inhibitory activity of flaxseed oil against CdCl 2 induced liver and kidney damage: Histopathology, genotoxicity, and gene expression study. Toxicol Rep 2020; 7:1127-1137. [PMID: 32963966 PMCID: PMC7490469 DOI: 10.1016/j.toxrep.2020.08.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 08/21/2020] [Accepted: 08/23/2020] [Indexed: 02/06/2023] Open
Abstract
The present work evaluated the effect of flaxseed oil (FO) against toxicity induced by cadmium chloride (CdCl2) in the mouse liver and kidney. Male Swiss albino mice were treated with CdCl2 (4.5 mg/kg, intraperitoneally) with or without FO at three concentrations (4, 8, 12 mL/kg, orally) for two consecutive weeks. To analyze the effects of FO, we used the following techniques: (1) histopathological examination; (2) comet assay; (3) RT-PCR gene expression analysis of tumor necrosis factor (TNF-α) and tumor suppressor protein (p53); and (4) immunohistochemical analysis of caspase-9 protein expression. The gas chromatography-mass spectrometry results showed that FO had a high content of unsaturated fatty acids including, oleic acid, linolenic acid, and linoleic acid. Oral supplementation with FO (12 mL/kg) resulted in a normal histological appearance without alteration in the DNA integrity and gene expression of TNF-α, p53, and caspase-9 in liver and kidney tissues. As expected, CdCl2 remarkably induced loss of histological integrity, increased DNA comet formation, increased TNF-α and p53 mRNA expression levels and increased the immunoreactivity of caspase-9 expression. When FO was given before administration of CdCl2, these histopathological defects were reversed; necrosis, degeneration, inflammatory cell infiltration, hemorrhage, Kupffer cells, and pyknotic cells were all reduced. These histological improvements induced by FO were accompanied by reduced DNA breakage, downregulated mRNA expression of TNF-α and p53, and downregulated immunohistochemical expression of caspase-9 protein. In conclusion, FO and its constituents may act as signaling molecules and modify the expression of genes involved in proinflammatory cytokine production (TNF-α), cell cycle arrest (p53), and apoptosis (caspase-9), thereby improving biological activities and health.
Collapse
Affiliation(s)
- Kawthar A. Diab
- Genetics and Cytology Department, Genetic Engineering and Biotechnology Division, National Research Centre (NRC), 33 El-Bohouth Street, Dokki, Cairo, P.O. 12622, Egypt
| | - Noha E. Ibrahim
- Microbial Biotechnology Department, Genetic Engineering and Biotechnology Division, National Research Centre (NRC), 33 El-Bohouth Street, Dokki, Cairo, P.O. 12622, Egypt
| | - Maha A. Fahmy
- Genetics and Cytology Department, Genetic Engineering and Biotechnology Division, National Research Centre (NRC), 33 El-Bohouth Street, Dokki, Cairo, P.O. 12622, Egypt
| | - Emad M. Hassan
- Medicinal and Aromatic Plants Research Department, Pharmaceutical Industries Research Division, National Research Centre (NRC), 33 El-Bohouth St, Dokki, Cairo, P.O. 12622, Egypt
| | - Enayat A. Omara
- Pathology Department, Medical Research Division, National Research Centre (NRC), 33 El-Bohouth Street, Dokki, Cairo, P.O. 12622, Egypt
| |
Collapse
|
11
|
Souza ACF, de Paiva Coimbra JL, Ervilha LOG, Bastos DSS, Cossolin JFS, Santos EC, de Oliveira LL, Machado-Neves M. Arsenic induces dose-dependent structural and ultrastructural pathological remodeling in the heart of Wistar rats. Life Sci 2020; 257:118132. [PMID: 32710949 DOI: 10.1016/j.lfs.2020.118132] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 07/07/2020] [Accepted: 07/18/2020] [Indexed: 12/18/2022]
Abstract
AIM Arsenic, an environmental contaminant, represents a public health problem worldwide. Studies have shown its association with molecular mechanisms related to cardiomyocytes redox balance. However, the microstructure and ultrastructure of cardiac tissue, as well as the activity of its antioxidant defenses front of disturbances in the mineral bioavailability induced by arsenic are still scarce. Thus, the aim of this study was to evaluate if arsenic exposure might induce structural and ultrastructural damages in cardiac tissue, including pathological remodeling of the parenchyma and stroma. Moreover, its impact on micromineral distribution and antioxidant enzymes activity in heart tissue was also evaluated. MAIN METHODS Adult male Wistar rats were divided into three groups that received 0, 1 and 10 mg/L sodium arsenite in drinking water for eight weeks. The hearts were collected and subjected to structural and ultrastructural analysis, mineral microanalysis and antioxidant enzymes quantification. Functional markers of cardiac damages were evaluated using serum samples. KEY FINDINGS Arsenic exposure induced dose-dependent structural and ultrastructural remodeling of cardiac tissue, with parenchyma loss, increase of stroma components, collagen deposition, and pathological damages such as inflammation, sarcomere disorganization, mitochondria degeneration and myofilament dissociation. Moreover, this metalloid was bioaccumulated in the tissue affecting its micromineral content, which resulted in antioxidant imbalance and increased levels of oxidative stress and cardiac markers. SIGNIFICANCE Taken together, our findings indicate that the heart is a potential target to arsenic toxicity, and long-term exposure to this metalloid must be avoided, once it might induce several cardiac tissue pathologies.
Collapse
Affiliation(s)
| | | | | | | | | | - Eliziária Cardoso Santos
- Medicine School, Federal University of Jequitinhonha and Mucuri Valleys, Minas Gerais, Brazil; Postgraduate Program in Animal Biology, Federal University of Jequitinhonha and Mucuri Valleys, Minas Gerais, Brazil
| | | | | |
Collapse
|
12
|
Ma W, Wei S, Zhang B, Li W. Molecular Mechanisms of Cardiomyocyte Death in Drug-Induced Cardiotoxicity. Front Cell Dev Biol 2020; 8:434. [PMID: 32582710 PMCID: PMC7283551 DOI: 10.3389/fcell.2020.00434] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 05/08/2020] [Indexed: 01/08/2023] Open
Abstract
Homeostatic regulation of cardiomyocytes plays a crucial role in maintaining the normal physiological activity of cardiac tissue. Severe cardiotoxicity results in cardiac diseases including but not limited to arrhythmia, myocardial infarction and myocardial hypertrophy. Drug-induced cardiotoxicity limits or forbids further use of the implicated drugs. Such drugs that are currently available in the clinic include anti-tumor drugs (doxorubicin, cisplatin, trastuzumab, etc.), antidiabetic drugs (rosiglitazone and pioglitazone), and an antiviral drug (zidovudine). This review focused on cardiomyocyte death forms and related mechanisms underlying clinical drug-induced cardiotoxicity, including apoptosis, autophagy, necrosis, necroptosis, pryoptosis, and ferroptosis. The key proteins involved in cardiomyocyte death signaling were discussed and evaluated, aiming to provide a theoretical basis and target for the prevention and treatment of drug-induced cardiotoxicity in the clinical practice.
Collapse
Affiliation(s)
- Wanjun Ma
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Shanshan Wei
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Bikui Zhang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Wenqun Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| |
Collapse
|
13
|
Haybar H, Shahrabi S, Rezaeeyan H, Jodat H, Saki N. Strategies to inhibit arsenic trioxide-induced cardiotoxicity in acute promyelocytic leukemia. J Cell Physiol 2019; 234:14500-14506. [PMID: 30770558 DOI: 10.1002/jcp.28292] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 12/19/2018] [Accepted: 12/20/2018] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Arsenic trioxide (ATO) is a drug commonly used for the treatment of acute promyelocytic leukemia (APL). Although ATO has been shown to cause significant improvement in patients, it is associated with serious side effects, which sometimes lead to the patient's death. In this review paper, we examine the reports of ATO-induced cardiotoxicity in APL patients and evaluate the strategies to reduce the incidence of such toxicity. METHODS The key search terms were "arsenic trioxide," "acute promyelocytic leukemia," "cardiotoxicity," "molecular pathway," and "biomarker." RESULTS Studies have indicated the involvement of several molecular pathways in ATO-induced cardiotoxicity. These pathways increase the production of reactive oxygen species by interfering with intracellular calcium homeostasis as well as impairing the transfer of calcium into endoplasmic reticulum and mitochondria. On the other hand, increasing or decreasing expressions of some microRNAs (miRs) have been shown to play a role in cardiotoxicity. CONCLUSION Finally, it can be stated that given the essential role of molecular pathways in cardiotoxicity and considering the fact these pathways impair the regulation of miRs expression, identification of molecular pathways involved in ATO-induced cardiotoxicity aimed at targeting miRs could be a new therapeutic strategy to prevent cardiotoxicity.
Collapse
Affiliation(s)
- Habib Haybar
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Saeid Shahrabi
- Department of Biochemistry and Hematology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Hadi Rezaeeyan
- Thalassemia and Hemoglobinopathy Research Center, Research Institute of Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hosein Jodat
- Thalassemia and Hemoglobinopathy Research Center, Research Institute of Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Najmaldin Saki
- Thalassemia and Hemoglobinopathy Research Center, Research Institute of Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
14
|
Iqubal A, Iqubal MK, Sharma S, Ansari MA, Najmi AK, Ali SM, Ali J, Haque SE. Molecular mechanism involved in cyclophosphamide-induced cardiotoxicity: Old drug with a new vision. Life Sci 2018; 218:112-131. [PMID: 30552952 DOI: 10.1016/j.lfs.2018.12.018] [Citation(s) in RCA: 185] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 11/08/2018] [Accepted: 12/10/2018] [Indexed: 12/20/2022]
Abstract
Cyclophosphamide (CP) is an important anticancer drug which belongs to the class of alkylating agent. Cyclophosphamide is mostly used in bone marrow transplantation, rheumatoid arthritis, lupus erythematosus, multiple sclerosis, neuroblastoma and other types of cancer. Dose-related cardiotoxicity is a limiting factor for its use. CP-induced cardiotoxicity ranges from 7 to 28% and mortality ranges from 11 to 43% at the therapeutic dose of 170-180 mg/kg, i.v. CP undergoes hepatic metabolism that results in the production of aldophosphamide. Aldophosphamide decomposes into phosphoramide mustard & acrolein. Phosphoramide is an active neoplastic agent, and acrolein is a toxic metabolite which acts on the myocardium and endothelial cells. This is the first review article that talks about cyclophosphamide-induced cardiotoxicity and the different signaling pathways involved in its pathogenicity. Based on the available literature, CP is accountable for cardiomyocytes energy pool alteration by affecting the heart fatty acid binding proteins (H-FABP). CP has been found associated with cardiomyocytes apoptosis, inflammation, endothelial dysfunction, calcium dysregulation, endoplasmic reticulum damage, and mitochondrial damage. Molecular mechanism of cardiotoxicity has been discussed in detail through crosstalk of Nrf2/ARE, Akt/GSK-3β/NFAT/calcineurin, p53/p38MAPK, NF-kB/TLR-4, and Phospholamban/SERCA-2a signaling pathway. Based on the available literature we support the fact that metabolites of CP are responsible for cardiotoxicity due to depletion of antioxidants/ATP level, altered contractility, damaged endothelium and enhanced pro-inflammatory/pro-apoptotic activities resulting into cardiomyopathy, myocardial infarction, and heart failure. Dose adjustment, elimination/excretion of acrolein and maintenance of endogenous antioxidant pool could be the therapeutic approach to mitigate the toxicities.
Collapse
Affiliation(s)
- Ashif Iqubal
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Mohammad Kashif Iqubal
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Sumit Sharma
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Mohd Asif Ansari
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Abul Kalam Najmi
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Syed Mansoor Ali
- Department of Biosciences, Jamia Millia Islamia,110025 New Delhi, India
| | - Javed Ali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Syed Ehtaishamul Haque
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
15
|
Zhang JY, Wang M, Wang RY, Sun X, Du YY, Ye JX, Sun GB, Sun XB. Salvianolic Acid A Ameliorates Arsenic Trioxide-Induced Cardiotoxicity Through Decreasing Cardiac Mitochondrial Injury and Promotes Its Anticancer Activity. Front Pharmacol 2018; 9:487. [PMID: 29867492 PMCID: PMC5954107 DOI: 10.3389/fphar.2018.00487] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 04/24/2018] [Indexed: 11/18/2022] Open
Abstract
Arsenic trioxide (ATO) is used as a therapeutic agent in the treatment of acute promyelocytic leukemia (APL). The therapeutic use of arsenic is limited due to its severe cardiovascular side effects. The cardio-protective effect of salvianolic acid A (Sal A) against ATO cardiotoxicity has been reported. However, the distinct role of the mitochondria in the cardio-protection of Sal A is not understood. The aim of this study was to determine whether Sal A preconditioning protects against ATO-induced heart injury by maintaining cardiac mitochondrial function and biogenesis. For the in vivo study, BALB/c mice were treated with ATO and/or Sal A. For the in vitro study, we determined the effects of ATO and/or Sal A in H9c2 cardiomyocytes. Our results showed that ATO induced mitochondrial structural damage, abnormal mitochondrial permeability transition pore (mPTP) opening, overproduction of mitochondrial reactive oxygen species (ROS), and decreased the ATP content. Sal A pretreatment alleviated the ATO-induced mitochondrial structural and functional damage. In this study, ATO decreased the expression level of the peroxisome proliferator activator receptor gamma-coactivator 1 (PGC-1α) and disrupted the normal division and fusion of mitochondria. Sal A pretreatment improved the dynamic balance of the damaged mitochondrial biogenesis. Moreover, the combination treatment of Sal A and ATO significantly enhanced the ATO-induced cytotoxicity of SGC7901, HepaRG, K562 and HL60 cells in vitro. These results indicated that Sal A protects the heart from ATO-induced injury, which correlates with the modulation of mitochondrial function, and the maintenance of normal mitochondrial biogenesis.
Collapse
Affiliation(s)
- Jing-Yi Zhang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China.,Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China.,Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Beijing, China
| | - Min Wang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China.,Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China.,Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Beijing, China
| | - Rui-Ying Wang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China.,Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China.,Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Beijing, China
| | - Xiao Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China.,Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China.,Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Beijing, China
| | - Yu-Yang Du
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China.,Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China.,Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Beijing, China
| | - Jing-Xue Ye
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China.,Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China.,Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Beijing, China
| | - Gui-Bo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China.,Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China.,Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Beijing, China
| | - Xiao-Bo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China.,Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China.,Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Beijing, China
| |
Collapse
|
16
|
Vineetha RC, Binu P, Arathi P, Nair RH. L-ascorbic acid and α-tocopherol attenuate arsenic trioxide-induced toxicity in H9c2 cardiomyocytes by the activation of Nrf2 and Bcl2 transcription factors. Toxicol Mech Methods 2018; 28:353-360. [PMID: 29297235 DOI: 10.1080/15376516.2017.1422578] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Arsenic trioxide (As2O3) is a potent drug for the treatment of acute promyelocytic leukemia (APL) and has achieved remarkable remissions in patients. Unfortunately, clinical reports have shown that the treatment is associated with cardiotoxicity. Many efforts have been made to mitigate drug-mediated cardiac damage using naturally occurring antioxidant compounds possessing free radical scavenging activity. The present investigation aims to explore protective role of L-ascorbic acid (L-AA) and α-tocopherol (α-TOC) from As2O3-induced oxidative stress in H9c2 cardiomyocytes through the evaluation of Nrf2 (nuclear factor erythroid 2-related factor 2) and Bcl-2 (B-cell lymphoma 2) transcription factors. The in vitro study was conducted using H9c2 cardiomyocytes. The evaluation of total antioxidant capacity, mitochondrial membrane potential, cellular calcium concentration and reactive oxygen species generation was performed. Oxidative stress (Nrf2) and anti-apoptotic (Bcl2) signaling indicators were measured by reverse transcriptase polymerase chain reaction. A depletion of the total antioxidant capacity and mitochondrial transmembrane potential were observed in As2O3-treated cardiomyocytes. In addition, the cellular calcium concentration and ROS generation were found to be increased on treatment with As2O3 with the alterations in the activity of transcription factors, Nrf2 and Bcl2. Co-treatment of antioxidant vitamins with As2O3 resulted in a significant reversal of oxidative stress and alteration on the antioxidant defense through the activation of Nrf2 and Bcl2. L-AA and α-TOC alleviates As2O3-induced oxidative stress in cardiac cells by activating Nrf2 and Bcl2 transcription factors that results in increased cell survival and prevents apoptosis.
Collapse
Affiliation(s)
| | - Prakash Binu
- a School of Biosciences , Mahatma Gandhi University , Kottayam , Kerala , India
| | - Pettamanna Arathi
- a School of Biosciences , Mahatma Gandhi University , Kottayam , Kerala , India
| | | |
Collapse
|
17
|
Pace C, Dagda R, Angermann J. Antioxidants Protect against Arsenic Induced Mitochondrial Cardio-Toxicity. TOXICS 2017; 5:toxics5040038. [PMID: 29206204 PMCID: PMC5750566 DOI: 10.3390/toxics5040038] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 11/29/2017] [Accepted: 12/01/2017] [Indexed: 12/17/2022]
Abstract
Arsenic is a potent cardiovascular toxicant associated with numerous biomarkers of cardiovascular diseases in exposed human populations. Arsenic is also a carcinogen, yet arsenic trioxide is used as a therapeutic agent in the treatment of acute promyelotic leukemia (APL). The therapeutic use of arsenic is limited due to its severe cardiovascular side effects. Many of the toxic effects of arsenic are mediated by mitochondrial dysfunction and related to arsenic's effect on oxidative stress. Therefore, we investigated the effectiveness of antioxidants against arsenic induced cardiovascular dysfunction. A growing body of evidence suggests that antioxidant phytonutrients may ameliorate the toxic effects of arsenic on mitochondria by scavenging free radicals. This review identifies 21 antioxidants that can effectively reverse mitochondrial dysfunction and oxidative stress in cardiovascular cells and tissues. In addition, we propose that antioxidants have the potential to improve the cardiovascular health of millions of people chronically exposed to elevated arsenic concentrations through contaminated water supplies or used to treat certain types of leukemias. Importantly, we identify conceptual gaps in research and development of new mito-protective antioxidants and suggest avenues for future research to improve bioavailability of antioxidants and distribution to target tissues in order reduce arsenic-induced cardiovascular toxicity in a real-world context.
Collapse
Affiliation(s)
- Clare Pace
- Department of Environmental Science and Health, University of Nevada, Reno, NV 89557, USA.
| | - Ruben Dagda
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA.
| | - Jeff Angermann
- School of Community Health Sciences, University of Nevada, Reno, NV 89557, USA.
| |
Collapse
|
18
|
Chen H, Gu S, Dai H, Li X, Zhang Z. Dihydroartemisinin Sensitizes Human Lung Adenocarcinoma A549 Cells to Arsenic Trioxide via Apoptosis. Biol Trace Elem Res 2017; 179:203-212. [PMID: 28261759 DOI: 10.1007/s12011-017-0975-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Accepted: 02/15/2017] [Indexed: 11/29/2022]
Abstract
Recent studies have shown that arsenic trioxide (ATO) is an effective anti-cancer drug for treatment of acute promyelocytic leukemia and other types of human cancer. However, we have found that lung cancer cells constantly develop a high level of resistance to ATO. In this study, we have explored a possibility of combination of dihydroartemisinin (DHA) and ATO treatments to reduce ATO resistance of lung cancer cells. We determined the combinatory effects of DHA and ATO on cytotoxicity of human lung adenocarcinoma (A549) cells. We showed that co-exposure to DHA and ATO of A549 cells synergistically increased the cytotoxicity and apoptotic cell death in the cells. We found that the synergistic effect of DHA and ATO in promoting apoptosis mainly resulted from increased cellular level of reactive oxygen species (ROS) and DNA damage. ATO alone only exerted moderate growth inhibitory effects on A549 cells. The results indicate that DHA can significantly sensitize ATO-induced cytotoxicity of A549 lung cancer cells through apoptosis mediated by ROS-induced DNA damage. Interestingly, we found that the combinatory treatment of DHA and ATO did not result in significant adverse effects in normal human bronchial epithelial (HBE) cells. Our results further provide evidence for the potential application of combinatory effects of DHA and ATO as a safe therapy for human lung cancer.
Collapse
Affiliation(s)
- Hongyu Chen
- Department of Environmental Health and Occupational Medicine, West China School of Public Health, Sichuan University, No. 16, Section 3, Renmin Nan Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Shiyan Gu
- Department of Environmental Health and Occupational Medicine, West China School of Public Health, Sichuan University, No. 16, Section 3, Renmin Nan Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Huangmei Dai
- Department of Environmental Health and Occupational Medicine, West China School of Public Health, Sichuan University, No. 16, Section 3, Renmin Nan Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xinyang Li
- Department of Environmental Health and Occupational Medicine, West China School of Public Health, Sichuan University, No. 16, Section 3, Renmin Nan Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Zunzhen Zhang
- Department of Environmental Health and Occupational Medicine, West China School of Public Health, Sichuan University, No. 16, Section 3, Renmin Nan Road, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
19
|
Gu S, Lai Y, Chen H, Liu Y, Zhang Z. miR-155 mediates arsenic trioxide resistance by activating Nrf2 and suppressing apoptosis in lung cancer cells. Sci Rep 2017; 7:12155. [PMID: 28939896 PMCID: PMC5610328 DOI: 10.1038/s41598-017-06061-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 06/06/2017] [Indexed: 12/16/2022] Open
Abstract
Arsenic trioxide (ATO) resistance is a challenging problem in chemotherapy. However, the underlying mechanisms remain to be elucidated. In this study, we identified a high level of expression of miR-155 in a human lung adenocarcinoma A549R cell line that is highly resistant to ATO. We showed that the high level of miR-155 was associated with increased levels of cell survival, colony formation, cell migration and decreased cellular apoptosis, and this was mediated by high levels of Nrf2, NAD(P)H quinone oxidoreductase 1 (NQO1), heme oxygenase-1 (HO-1) and a high ratio of Bcl-2/Bax. Overexpression of the miR-155 mimic in A549R cells resulted in increased levels of colony formation and cell migration as well as reduced apoptosis along with increased Nrf2, NQO1 and HO-1. In contrast, silencing of miR-155 expression with its inhibitor in the cells, significantly decreased the cellular levels of Nrf2, NQO1 and HO-1 as well as the ratio of Bcl-2/Bax. This subsequently reduced the level of colony formation and cell migration facilitating ATO-induced apoptosis. Our results indicate that miR-155 mediated ATO resistance by upregulating the Nrf2 signaling pathway, but downregulating cellular apoptosis in lung cancer cells. Our study provides new insights into miR-155-mediated ATO resistance in lung cancer cells.
Collapse
Affiliation(s)
- Shiyan Gu
- Department of Environmental Health and Occupational Medicine, West China School of Public Health, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yanhao Lai
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida, 33199, USA
| | - Hongyu Chen
- Department of Environmental Health and Occupational Medicine, West China School of Public Health, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yuan Liu
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida, 33199, USA. .,Biochemistry Ph.D. Program, Florida International University, Miami, Florida, 33199, USA. .,Biomolecular Sciences Institute, Florida International University, Miami, Florida, 33199, USA.
| | - Zunzhen Zhang
- Department of Environmental Health and Occupational Medicine, West China School of Public Health, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
20
|
Varghese MV, Abhilash M, Alex M, Paul MVS, Prathapan A, Raghu KG, Nair RH. Attenuation of arsenic trioxide induced cardiotoxicity through flaxseed oil in experimental rats. Redox Rep 2017; 22:346-352. [PMID: 28209094 DOI: 10.1080/13510002.2017.1289313] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
OBJECTIVES Arsenic trioxide (As2O3) is a potent drug for acute promyelocytic leukaemia, but its clinical trials are allied with some serious adverse events mainly cardiac functional abnormalities. So the objective of our investigation is to identify the cardioprotective action of flaxseed oil (FSO), a natural compound against As2O3 induced cardiotoxicity. METHODS Male wistar rats were treated with As2O3 (4 mg/kg) to induce cardiotoxicity. FSO (250 and 500 mg/kg) was given in combination with As2O3 for evaluating its cardioprotective efficacy. RESULTS Treatment with As2O3 resulted in deposition of arsenic in heart tissue, increased cardiac marker enzymes release, lipid peroxidation (LPO), oxidative insults and pathological damages in the heart. Co-treatment with FSO (500 mg/kg) significantly reduced the arsenic accumulation, cardiac marker enzymes, LPO and cardiac structural alterations. FSO treatment significantly improved cardiac glutathione content, antioxidant enzymes and reduced the pathological damages in cardiac tissue. Gas chromatographic-mass spectrometry analysis revealed that the major fatty acid content in the FSO is alpha-linolenic acid, which has a strong milieu in cardiac health. CONCLUSION The results of the current investigation suggested that FSO is an effective agent in reducing arsenic-induced cardiac toxicity and can be used as an adjunct/dietary supplement for the cancer patients on As2O3 therapy.
Collapse
Affiliation(s)
| | - M Abhilash
- a School of Biosciences , Mahatma Gandhi University , Kottayam , India
| | - Manju Alex
- a School of Biosciences , Mahatma Gandhi University , Kottayam , India
| | - M V Sauganth Paul
- a School of Biosciences , Mahatma Gandhi University , Kottayam , India
| | - A Prathapan
- b Biochemistry and Molecular Mechanism Laboratory, Agroprocessing and Technology Division , CSIR-National Institute for Interdisciplinary Science and Technology (NIIST) , Trivandrum , India
| | - K G Raghu
- b Biochemistry and Molecular Mechanism Laboratory, Agroprocessing and Technology Division , CSIR-National Institute for Interdisciplinary Science and Technology (NIIST) , Trivandrum , India
| | | |
Collapse
|
21
|
Is LukS-PV a novel experimental therapy for leukemia? Gene 2016; 600:44-47. [PMID: 27916717 DOI: 10.1016/j.gene.2016.11.044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 11/17/2016] [Accepted: 11/30/2016] [Indexed: 12/15/2022]
Abstract
Although the studies on the pathogenesis and prognosis of leukemia have made revolutionary progress, the long-term survival remains unsatisfactory. Alternative techniques are being developed to target leukemia. Several decades after researchers' work, a variety of bacteria toxins are being explored as potential anti-leukemia agents, either to provide direct effects or to deliver therapeutic proteins to leukemia. LukS-PV, a component of Panton-Valentine Leukocidin secreted by S. aureus, has been tested in acute myeloid leukemia as a novel experimental strategy. Further researches about the targeting mechanisms of LukS-PV are required to make it a complete therapeutic approach for leukemia treatment. The function of this article is to provide clinicians and experimentalists with a chronological and comprehensive appraisal of use of LukS-PV as an experimental strategy for leukemia therapy.
Collapse
|