1
|
Tanwar SS, Dwivedi S, Khan S, Sharma S. Cardiomyopathies and a brief insight into DOX-induced cardiomyopathy. Egypt Heart J 2025; 77:29. [PMID: 40064787 PMCID: PMC11893974 DOI: 10.1186/s43044-025-00628-0] [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: 12/27/2024] [Accepted: 02/23/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Cardiomyopathy is a heterogeneous group of myocardial disorders characterized by structural and functional abnormalities of the heart muscle. It is classified into primary (genetic, mixed, or acquired) and secondary categories, resulting in various phenotypes including dilated, hypertrophic, and restrictive patterns. Hypertrophic cardiomyopathy, the most common primary form, can cause exertional dyspnea, presyncope, and sudden cardiac death. Dilated cardiomyopathy typically presents with heart failure symptoms, while restrictive cardiomyopathy is rarer and often associated with systemic diseases. Diagnosis involves a comprehensive evaluation including history, physical examination, electrocardiography, and echocardiography. Treatment options range from pharmacotherapy and lifestyle modifications to implantable cardioverter-defibrillators and heart transplantation in refractory cases. MAIN BODY Anthracyclines, particularly doxorubicin, have emerged as crucial components in cancer treatment, demonstrating significant antitumor activity across various malignancies. These drugs have become standard in numerous chemotherapy regimens, improving patient outcomes. However, their use is associated with severe cardiotoxicity, including cardiomyopathy and heart failure. The mechanisms of anthracycline action and toxicity are complex, involving DNA damage, iron-mediated free radical production, and disruption of cardiovascular homeostasis. Doxorubicin-induced cardiomyopathy (DIC) is a severe complication of cancer treatment with a poor prognosis and limited effective treatments. The pathophysiology of DIC involves multiple mechanisms, including oxidative stress, inflammation, mitochondrial damage, and calcium homeostasis disorder. Despite extensive research, no effective treatment for established DIC is currently available. Dexrazoxane is the only FDA-approved protective agent, but it has limitations. Recent studies have explored various potential therapeutic approaches, including natural drugs, endogenous substances, new dosage forms, and herbal medicines. However, the lack of experimental models incorporating pre-existing cancer limits the understanding of DIC pathophysiology and treatment efficacy. CONCLUSION Cardiomyopathy, whether primary or secondary, poses a significant clinical challenge due to its varying etiologies and poor prognosis in advanced stages. Anthracycline-induced cardiomyopathy is a severe complication of chemotherapy, with doxorubicin being a notable contributor. Despite advancements in cancer therapies, the cardiotoxic effects of anthracyclines necessitate further investigation into effective preventive strategies and therapeutic interventions to improve patient outcomes.
Collapse
Affiliation(s)
| | - Sumeet Dwivedi
- Acropolis Institute of Pharmaceutical Education and Research, Indore, India
| | - Sheema Khan
- The University of Texas Rio Grande Valley, Edinburg, US
| | - Seema Sharma
- Shri Vaishnav Vidyapeeth Vishwadvidyalaya, Indore, India.
| |
Collapse
|
2
|
El-Serafi I, Steele S. Cyclophosphamide Pharmacogenomic Variation in Cancer Treatment and Its Effect on Bioactivation and Pharmacokinetics. Adv Pharmacol Pharm Sci 2024; 2024:4862706. [PMID: 38966316 PMCID: PMC11223907 DOI: 10.1155/2024/4862706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 06/06/2024] [Accepted: 06/11/2024] [Indexed: 07/06/2024] Open
Abstract
Cyclophosphamide (Cy) is a prodrug that is mainly bioactivated by cytochrome P450 (CYP) 2B6 enzyme. Several other enzymes are also involved in its bioactivation and affect its kinetics. Previous studies have shown the effect of the enzymes' genetic polymorphisms on Cy kinetics and its clinical outcome. These results were controversial primarily because of the involvement of several interacting enzymes in the Cy metabolic pathway, which can also be affected by several clinical factors as well as other drug interactions. In this review article, we present the effect of CYP2B6 polymorphisms on Cy kinetics since it is the main bioactivating enzyme, as well as discussing all previously reported enzymes and clinical factors that can alter Cy efficacy. Additionally, we present explanations for key Cy side effects related to the nature and site of its bioactivation. Finally, we discuss the role of busulphan in conditioning regimens in the Cy metabolic pathway as a clinical example of drug-drug interactions involving several enzymes. By the end of this article, our aim is to have provided a comprehensive summary of Cy pharmacogenomics and the effect on its kinetics. The utility of these findings in the development of new strategies for Cy personalized patient dose adjustment will aid in the future optimization of patient specific Cy dosages and ultimately in improving clinical outcomes. In conclusion, CYP2B6 and several other enzyme polymorphisms can alter Cy kinetics and consequently the clinical outcomes. However, the precise quantification of Cy kinetics in any individual patient is complex as it is clearly under multifactorial genetic control. Additionally, other clinical factors such as the patient's age, diagnosis, concomitant medications, and clinical status should also be considered.
Collapse
Affiliation(s)
- Ibrahim El-Serafi
- Basic Medical Sciences DepartmentCollege of MedicineAjman University, Ajman, UAE
- Department of Hand Surgery, and Plastic Surgery and BurnsLinköping University Hospital, Linkoöping, Sweden
| | - Sinclair Steele
- Pathological Sciences DepartmentCollege of MedicineAjman University, Ajman, UAE
| |
Collapse
|
3
|
Gu B, Han K, Cao H, Huang X, Li X, Mao M, Zhu H, Cai H, Li D, He J. Heart-on-a-chip systems with tissue-specific functionalities for physiological, pathological, and pharmacological studies. Mater Today Bio 2024; 24:100914. [PMID: 38179431 PMCID: PMC10765251 DOI: 10.1016/j.mtbio.2023.100914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 12/08/2023] [Accepted: 12/12/2023] [Indexed: 01/06/2024] Open
Abstract
Recent advances in heart-on-a-chip systems hold great promise to facilitate cardiac physiological, pathological, and pharmacological studies. This review focuses on the development of heart-on-a-chip systems with tissue-specific functionalities. For one thing, the strategies for developing cardiac microtissues on heart-on-a-chip systems that closely mimic the structures and behaviors of the native heart are analyzed, including the imitation of cardiac structural and functional characteristics. For another, the development of techniques for real-time monitoring of biophysical and biochemical signals from cardiac microtissues on heart-on-a-chip systems is introduced, incorporating cardiac electrophysiological signals, contractile activity, and biomarkers. Furthermore, the applications of heart-on-a-chip systems in intelligent cardiac studies are discussed regarding physiological/pathological research and pharmacological assessment. Finally, the future development of heart-on-a-chip toward a higher level of systematization, integration, and maturation is proposed.
Collapse
Affiliation(s)
- Bingsong Gu
- State Key Laboratory for Manufacturing System Engineering, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Innovation Platform (Center) for Industry-Education Integration of Medical Technology, Xi'an Jiaotong University, China
| | - Kang Han
- State Key Laboratory for Manufacturing System Engineering, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Innovation Platform (Center) for Industry-Education Integration of Medical Technology, Xi'an Jiaotong University, China
| | - Hanbo Cao
- Shaanxi Provincial Institute for Food and Drug Control, Xi’ an, 710065, China
| | - Xinxin Huang
- State Key Laboratory for Manufacturing System Engineering, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Innovation Platform (Center) for Industry-Education Integration of Medical Technology, Xi'an Jiaotong University, China
| | - Xiao Li
- State Key Laboratory for Manufacturing System Engineering, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Innovation Platform (Center) for Industry-Education Integration of Medical Technology, Xi'an Jiaotong University, China
| | - Mao Mao
- State Key Laboratory for Manufacturing System Engineering, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Innovation Platform (Center) for Industry-Education Integration of Medical Technology, Xi'an Jiaotong University, China
| | - Hui Zhu
- State Key Laboratory for Manufacturing System Engineering, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Innovation Platform (Center) for Industry-Education Integration of Medical Technology, Xi'an Jiaotong University, China
| | - Hu Cai
- Shaanxi Provincial Institute for Food and Drug Control, Xi’ an, 710065, China
| | - Dichen Li
- State Key Laboratory for Manufacturing System Engineering, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Innovation Platform (Center) for Industry-Education Integration of Medical Technology, Xi'an Jiaotong University, China
| | - Jiankang He
- State Key Laboratory for Manufacturing System Engineering, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Innovation Platform (Center) for Industry-Education Integration of Medical Technology, Xi'an Jiaotong University, China
| |
Collapse
|
4
|
Zhong Y, Zhou L, Wang H, Lin S, Liu T, Kong X, Xiao G, Gao H. Kindlin-2 maintains liver homeostasis by regulating GSTP1-OPN-mediated oxidative stress and inflammation in mice. J Biol Chem 2024; 300:105601. [PMID: 38159860 PMCID: PMC10831259 DOI: 10.1016/j.jbc.2023.105601] [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: 09/11/2023] [Revised: 12/16/2023] [Accepted: 12/18/2023] [Indexed: 01/03/2024] Open
Abstract
Hepatocyte plays a principal role in preserving integrity of the liver homeostasis. Our recent study demonstrated that Kindlin-2, a focal adhesion protein that activates integrins and regulates cell-extracellular matrix interactions, plays an important role in regulation of liver homeostasis by inhibiting inflammation pathway; however, the molecular mechanism of how Kindlin-2 KO activates inflammation is unknown. Here, we show that Kindlin-2 loss largely downregulates the antioxidant glutathione-S-transferase P1 in hepatocytes by promoting its ubiquitination and degradation via a mechanism involving protein-protein interaction. This causes overproduction of intracellular reactive oxygen species and excessive oxidative stress in hepatocytes. Kindlin-2 loss upregulates osteopontin in hepatocytes partially because of upregulation of reactive oxygen species and consequently stimulates overproduction of inflammatory cytokines and infiltration in liver. The molecular and histological deteriorations caused by Kindlin-2 deficiency are markedly reversed by systemic administration of an antioxidant N-acetylcysteine in mice. Taken together, Kindlin-2 plays a pivotal role in preserving integrity of liver function.
Collapse
Affiliation(s)
- Yiming Zhong
- Shanghai Key Laboratory of Metabolic Remodeling and Health, State Key Laboratory of Genetic Engineering, Institute of Metabolism and Integrative Biology, School of Life Sciences, Jinshan Hospital, Fudan University, Shanghai, China; Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, China
| | - Liang Zhou
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Hui Wang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, State Key Laboratory of Genetic Engineering, Institute of Metabolism and Integrative Biology, School of Life Sciences, Jinshan Hospital, Fudan University, Shanghai, China
| | - Sixiong Lin
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Department of Spinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Tiemin Liu
- Shanghai Key Laboratory of Metabolic Remodeling and Health, State Key Laboratory of Genetic Engineering, Institute of Metabolism and Integrative Biology, School of Life Sciences, Jinshan Hospital, Fudan University, Shanghai, China.
| | - Xingxing Kong
- Shanghai Key Laboratory of Metabolic Remodeling and Health, State Key Laboratory of Genetic Engineering, Institute of Metabolism and Integrative Biology, School of Life Sciences, Jinshan Hospital, Fudan University, Shanghai, China.
| | - Guozhi Xiao
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, China.
| | - Huanqing Gao
- Shanghai Key Laboratory of Metabolic Remodeling and Health, State Key Laboratory of Genetic Engineering, Institute of Metabolism and Integrative Biology, School of Life Sciences, Jinshan Hospital, Fudan University, Shanghai, China; Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
5
|
İpek E, Hesapçıoğlu M, Karaboğa M, Avcı H. Selenium protection from DNA damage and regulation of apoptosis signaling following cyclophosphamide induced cardiotoxicity in rats. Biotech Histochem 2023; 98:534-542. [PMID: 37695070 DOI: 10.1080/10520295.2023.2253424] [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] [Indexed: 09/12/2023] Open
Abstract
We investigated the mechanism of the cardioprotective effect of selenium (Se) against cyclophosphamide (CPA) induced cardiotoxicity in rats. We divided 24 female Wistar albino rats into four groups. The control group was injected intraperitoneally (i.p.) with normal saline. The CPA group was injected i.p. with 200 mg/kg CPA. The Se group was injected i.p. with 1 mg/kg Se. The CPA + Se group was injected i.p. with 200 mg/kg CPA and 1 mg/kg Se. Rats were euthanized 24 h after injection and heart tissues were harvested. Histopathological examination revealed reduced severity of myocardial lesions in the CPA + Se group compared to CPA induced cardiotoxicity of the CPA group; this finding was confirmed by increased immunoreactivity of cardiac troponin-I (cTn-I) in the CPA + Se group compared to decreased cTn-I immunoreactivity in the CPA group. Administration of CPA increased the immunoreactivity of phosphorylated histone-2AX (γH2AX). Se reduced the CPA induced increase in γH2AX immunoreactivity. Se administration reversed the CPA induced increase of Bax and decrease of Bcl2 gene expressions. Our findings suggest that Se is cardioprotective by reducing DNA damage and regulating the genes responsible for apoptosis caused by CPA in rats.
Collapse
Affiliation(s)
- Emrah İpek
- Department of Pathology, Faculty of Veterinary Medicine, Aydın Adnan Menderes University, Aydın, Turkey
| | - Mehmet Hesapçıoğlu
- Department of Pathology, Faculty of Veterinary Medicine, Aydın Adnan Menderes University, Aydın, Turkey
| | - Mehmet Karaboğa
- Department of Pathology, Faculty of Veterinary Medicine, Aydın Adnan Menderes University, Aydın, Turkey
| | - Hamdi Avcı
- Department of Pathology, Faculty of Veterinary Medicine, Aydın Adnan Menderes University, Aydın, Turkey
| |
Collapse
|
6
|
Xiong J, Ding B, Zhu W, Xu L, Yu S. Exosomes from Adipose-Derived Mesenchymal Stem Cells Protect Against Cyclophosphamide-Induced Cardiotoxicity in Rats. Int Heart J 2023; 64:935-944. [PMID: 37778997 DOI: 10.1536/ihj.23-201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
A certain dosage of cyclophosphamide (CYP) in clinical applications contributes to severe cardiotoxicity. Herein, this study explored the impact of adipose-derived mesenchymal stem cell (AdMSC)-exosomes (Exos) on CYP-induced cardiotoxicity.AdMSCs and AdMSCs-Exos were isolated and identified. CYP was utilized for developing a cardiotoxicity rat model, after which blood was collected and then the serum contents of cardiac injury-related indexes (creatine kinase-MB, lactate dehydrogenase, aspartate aminotransferase, and alkaline phosphatase) were detected with enzyme-linked immunosorbent assay kits. Oxidative stress (OS)-related indicators were measured with the corresponding kits. Myocardial pathological changes and collagen fibrosis were tested with hematoxylin-eosin and Masson staining, and apoptosis-related and autophagy-related proteins in rat cardiac tissues with immunohistochemistry and Western blot assays, respectively.AdMSCs and AdMSCs-Exos were successfully isolated. AdMSCs-Exos could target rat hearts. AdMSCs-Exos improved cardiac function and diminished the content of the cardiac injury-related indexes in CYP rats. In addition, AdMSCs-Exos reduced CYP-induced cardiac fibrosis, OS, apoptosis, and autophagy in rats.AdMSCs-Exos alleviated CYP-induced cardiotoxicity in rats via the repression of OS, apoptosis, and autophagy.
Collapse
Affiliation(s)
- Jianhua Xiong
- Department of General Medicine, Fuzhou First People's Hospital
| | - Binjun Ding
- Department of General Medicine, Fuzhou First People's Hospital
| | - Wei Zhu
- Department of Cardiovascular Medicine, Fuzhou First People's Hospital
| | - Lanlan Xu
- Department of General Medicine, Fuzhou First People's Hospital
| | - Songping Yu
- Department of Cardiology, Jiangxi Provincial People's Hospital
| |
Collapse
|
7
|
Chen H, Zhou H, Yang J, Wan H, He Y. Guhong injection mitigates myocardial ischemia/reperfusion injury by activating GST P to inhibit ASK1-JNK/p38 pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 109:154603. [PMID: 36610111 DOI: 10.1016/j.phymed.2022.154603] [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: 10/11/2022] [Revised: 11/30/2022] [Accepted: 12/12/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND Guhong injection (GHI), a novel compound preparation that is composed of a chemical drug, namely aceglutamide, and the aqueous extract of safflower (Carthamus tinctorius L.), exhibits extreme antioxidative, antiapoptotic, anti-inflammatory, and neuroprotective effects. Since oxidative stress, apoptosis, and inflammatory response are all the dominant mechanisms of myocardial ischemia/reperfusion (MI/R) injury, we probe into the protective mechanism of GHI on MI/R injury for the first time. METHODS In this research, we first employed molecular docking to determine whether three active ingredients in GHI, acetylglutamine (NAG), hydroxysafflor yellow A (HSYA), and syringin, possessed the potential activity to modulate the protein, glutathione S-transferase P (GST P). We further identified the protective effect of GHI on myocardial tissue with TTC staining, HE staining, TUNEL staining, and ELISA, and on H9c2 with flow cytometry and ELISA. We next explored whether the cardioprotective effect of GHI on left anterior descending ligation-reperfusion in rats and hypoxia/reoxygenation (H/R) in H9c2 cells was related to activate GST P to inhibit ASK1-JNK/p38 pathway via approaches of qRT-PCR and Western blot. RESULTS Results of molecular docking indicated that all three compounds spontaneously docked to GST P, among them the binding affinities of both HSYA and syringin to GST P were higher than NAG. In vivo, GHI reduced myocardial infarction size and mitigated myocardial pathological injury. In vitro, GHI enhanced cell viability and extenuated depolarization of mitochondrial membrane potential. In addition, the results of in vivo and in vitro studies demonstrated that the cardioprotection of GHI was associated with improving the mRNA and protein expression levels of GST P to modulate oxidative stress, and inhibiting the levels of mRNA expression and protein phosphorylation of ASK1, JNK, and p38. However, the suppressed effect of GHI on ASK1-JNK/p38 pathway was reversed by ethacrynic acid (EA, a GST inhibitor), indicating that the regulation of GHI on ASK1-JNK/p38 was related to the activity of GST P. Besides, the in vitro results of qRT-PCR and western-blot also certified that the inhibited JNK and p38 further reduced Bax expression and elevated Bcl-2 expression to reduce the expression of caspase-3 to exert anti-apoptosis effects. CONCLUSION Taken together, the cardioprotection of GHI mainly incarnated in activating GST P to relieve oxidation properties, thereby inhibiting ASK1-JNK/p38 pathway to suppress apoptosis.
Collapse
Affiliation(s)
- Haiyang Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Huifen Zhou
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Jiehong Yang
- School of Basic Medicine Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Haitong Wan
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| | - Yu He
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| |
Collapse
|
8
|
Singh SK, Mukerjee A, Gupta P, Kumar Tripathi A. Evaluation of Antigenotoxic Effect of Cinnamon Oil and Usnic Acid Blended Nanoemulsion on Swiss Albino Mice. BIONANOSCIENCE 2022. [DOI: 10.1007/s12668-021-00902-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
9
|
Dionísio F, Araújo AM, Duarte-Araújo M, Bastos MDL, Guedes de Pinho P, Carvalho F, Costa VM. Cardiotoxicity of cyclophosphamide's metabolites: an in vitro metabolomics approach in AC16 human cardiomyocytes. Arch Toxicol 2022; 96:653-671. [PMID: 35088106 DOI: 10.1007/s00204-021-03204-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 12/09/2021] [Indexed: 12/11/2022]
Abstract
Cyclophosphamide is a widely used anticancer and immunosuppressive prodrug that unfortunately causes severe adverse effects, including cardiotoxicity. Although the exact cardiotoxic mechanisms are not completely understood, a link between cyclophosphamide's pharmacologically active metabolites, namely 4-hydroxycyclophosphamide and acrolein, and the toxicity observed after the administration of high doses of the prodrug is likely. Therefore, the objective of this study is to shed light on the cardiotoxic mechanisms of cyclophosphamide and its main biotransformation products, through classic and metabolomics studies. Human cardiac proliferative and differentiated AC16 cells were exposed to several concentrations of the three compounds, determining their basic cytotoxic profile and preparing the next study, using subtoxic and toxic concentrations for morphological and biochemical studies. Finally, metabolomics studies were applied to cardiac cells exposed to subtoxic concentrations of the aforementioned compounds to determine early markers of damage. The cytotoxicity, morphological and biochemical assays showed that 4-hydroxycyclophosphamide and acrolein induced marked cardiotoxicity at µM concentrations (lower than 5 µM), being significantly lower than the ones observed for cyclophosphamide (higher than 2500 μM). Acrolein led to increased levels of ATP and total glutathione on proliferative cells at 25 µM, while no meaningful changes were observed in differentiated cells. Higher levels of carbohydrates and decreased levels of fatty acids and monoacylglycerols indicated a metabolic cardiac shift after exposure to cyclophosphamide's metabolites, as well as a compromise of precursor amino acids used in the synthesis of glutathione, seen in proliferative cells' metabolome. Overall, differences in cytotoxic mechanisms were observed for the two different cellular states used and for the three molecules, which should be taken into consideration in the study of cyclophosphamide cardiotoxic mechanisms.
Collapse
Affiliation(s)
- Flávio Dionísio
- UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira nº 228, 4050-313, Porto, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
| | - Ana Margarida Araújo
- UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira nº 228, 4050-313, Porto, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
| | - Margarida Duarte-Araújo
- LAQV/REQUIMTE, Department of Imuno-Physiology and Pharmacology, Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal
| | - Maria de Lourdes Bastos
- UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira nº 228, 4050-313, Porto, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
| | - Paula Guedes de Pinho
- UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira nº 228, 4050-313, Porto, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
| | - Félix Carvalho
- UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira nº 228, 4050-313, Porto, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
| | - Vera Marisa Costa
- UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira nº 228, 4050-313, Porto, Portugal.
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.
| |
Collapse
|
10
|
Queirós V, Azeiteiro UM, Barata C, Santos JL, Alonso E, Soares AMVM, Freitas R. Effects of the antineoplastic drug cyclophosphamide on the biochemical responses of the mussel Mytilus galloprovincialis under different temperatures. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 288:117735. [PMID: 34271515 DOI: 10.1016/j.envpol.2021.117735] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 07/02/2021] [Accepted: 07/04/2021] [Indexed: 06/13/2023]
Abstract
Cyclophosphamide (CP) is an antineoplastic drug widely used in chemotherapy treatments with high consumption rates and that has been detected in the aquatic environment. After being released into the aquatic environment, CP may cause adverse effects on aquatic organisms since antineoplastics are well-known cytotoxic, genotoxic, mutagenic and teratogenic drugs. Moreover, predicted environmental changes, such as the temperature rising, may alter the impacts caused by CP on organisms. Thus, the present study aimed to assess the effects caused by CP chronic exposure in the mussel Mytilus galloprovincialis, under actual and predicted warming scenarios. Organisms were exposed for 28 days to different concentrations of CP (10, 100, 500 and 1000 ng/L) at control (17 ± 1.0 °C) and increased (21 ± 1.0 °C) temperatures. Biochemical responses related to metabolic capacity, energy reserves, oxidative stress and neurotoxicity were assessed. The results showed that the organisms were able to maintain their metabolic capacity under all exposure conditions. However, their antioxidant defense mechanisms were activated mostly at higher CP concentrations being able to prevent cellular damage, even under the warming scenario. Overall, the present findings suggest that temperature rise may not alter the impacts of CP towards M. galloprovincialis.
Collapse
Affiliation(s)
- Vanessa Queirós
- Biology Department & CESAM, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Ulisses M Azeiteiro
- Biology Department & CESAM, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Carlos Barata
- Department of Environmental Chemistry, Institute of Environmental Assessment and Water Research, IDAEA-CSIC, Jordi Girona 18, 08034, Barcelona, Spain
| | - Juan Luis Santos
- Departamento de Química Analítica, Escuela Politécnica Superior, Universidad de Sevilla, Spain
| | - Esteban Alonso
- Departamento de Química Analítica, Escuela Politécnica Superior, Universidad de Sevilla, Spain
| | - Amadeu M V M Soares
- Biology Department & CESAM, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Rosa Freitas
- Biology Department & CESAM, University of Aveiro, 3810-193, Aveiro, Portugal.
| |
Collapse
|
11
|
Jin L, Lorkiewicz P, Xie Z, Bhatnagar A, Srivastava S, Conklin DJ. Acrolein but not its metabolite, 3-Hydroxypropylmercapturic acid (3HPMA), activates vascular transient receptor potential Ankyrin-1 (TRPA1): Physiological to toxicological implications. Toxicol Appl Pharmacol 2021; 426:115647. [PMID: 34271065 PMCID: PMC8343963 DOI: 10.1016/j.taap.2021.115647] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 11/17/2022]
Abstract
Acrolein, an electrophilic α,β-unsaturated aldehyde, is present in foods and beverages, and is a product of incomplete combustion, and thus, reaches high ppm levels in tobacco smoke and structural fires. Exposure to acrolein is linked with cardiopulmonary toxicity and cardiovascular disease risk. The hypothesis of this study is the direct effects of acrolein in isolated murine blood vessels (aorta and superior mesenteric artery, SMA) are transient receptor potential ankyrin-1 (TRPA1) dependent. Using isometric myography, isolated aorta and SMA were exposed to increasing levels of acrolein. Acrolein inhibited phenylephrine (PE)-induced contractions (approximately 90%) in aorta and SMA of male and female mice in a concentration-dependent (0.01-100 μM) manner. The major metabolite of acrolein, 3-hydroxypropylmercapturic acid (3HPMA), also relaxed PE-precontracted SMA. As the SMA was 20× more sensitive to acrolein than aorta (SMA EC50 0.8 ± 0.2 μM; aorta EC50 > 29.4 ± 4.4 μM), the mechanisms of acrolein-induced relaxation were studied in SMA. The potency of acrolein-induced relaxation was inhibited significantly by: 1) mechanically-impaired endothelium; 2) Nω-Nitro-L-arginine methyl ester hydrochloride (L-NAME); 3) guanylyl cyclase (GC) inhibitor (ODQ); and, 4) a TRPA1 antagonist (A967079). TRPA1 positive immunofluorescence was present in the endothelium. Compared with other known TRPA1 agonists, including allyl isothiocyanate (AITC), cinnamaldehyde, crotonaldehyde, and formaldehyde, acrolein stimulated a more potent TRPA1-dependent relaxation. Acrolein, at high concentration [100 μM], induced tension oscillations (spasms) independent of TRPA1 in precontracted SMA but not in aorta. In conclusion, acrolein is vasorelaxant at low levels (physiological) yet vasotoxic at high levels (toxicological).
Collapse
Affiliation(s)
- L Jin
- Christina Lee Brown Envirome Institute, University of Louisville, Louisville, KY, USA; Diabetes and Obesity Center, University of Louisville, Louisville, KY, USA; American Heart Association-Tobacco Regulation and Addiction Center, University of Louisville, Louisville, KY, USA
| | - P Lorkiewicz
- Christina Lee Brown Envirome Institute, University of Louisville, Louisville, KY, USA; Diabetes and Obesity Center, University of Louisville, Louisville, KY, USA; American Heart Association-Tobacco Regulation and Addiction Center, University of Louisville, Louisville, KY, USA
| | - Z Xie
- Christina Lee Brown Envirome Institute, University of Louisville, Louisville, KY, USA; Diabetes and Obesity Center, University of Louisville, Louisville, KY, USA
| | - A Bhatnagar
- Christina Lee Brown Envirome Institute, University of Louisville, Louisville, KY, USA; Diabetes and Obesity Center, University of Louisville, Louisville, KY, USA; American Heart Association-Tobacco Regulation and Addiction Center, University of Louisville, Louisville, KY, USA; Superfund Research Center, University of Louisville, Louisville, KY, USA
| | - S Srivastava
- Christina Lee Brown Envirome Institute, University of Louisville, Louisville, KY, USA; Diabetes and Obesity Center, University of Louisville, Louisville, KY, USA; American Heart Association-Tobacco Regulation and Addiction Center, University of Louisville, Louisville, KY, USA; Superfund Research Center, University of Louisville, Louisville, KY, USA
| | - D J Conklin
- Christina Lee Brown Envirome Institute, University of Louisville, Louisville, KY, USA; Diabetes and Obesity Center, University of Louisville, Louisville, KY, USA; American Heart Association-Tobacco Regulation and Addiction Center, University of Louisville, Louisville, KY, USA; Superfund Research Center, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
12
|
Zhang W, Gao J, Lu L, Bold T, Li X, Wang S, Chang Z, Chen J, Kong X, Zheng Y, Zhang M, Tang J. Intracellular GSH/GST antioxidants system change as an earlier biomarker for toxicity evaluation of iron oxide nanoparticles. NANOIMPACT 2021; 23:100338. [PMID: 35559839 DOI: 10.1016/j.impact.2021.100338] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 06/14/2021] [Accepted: 06/18/2021] [Indexed: 05/14/2023]
Abstract
Glutathione (GSH) and glutathione-S-transferases (GSTs) are two frontlines of cellular defense against both acute and chronic toxicity of xenobiotics-induced oxidative stress. The contribution of GSH and GST enzymes to signaling pathways and the regulation of GSH homeostasis play a central role in the detoxification of numerous environmental toxins and impurities. Iron oxide nanoparticles stemmed from traffic exhaust, steel manufacturing, or welding as a potential environmental pollution can lead to adverse respiratory outcomes and aggravate the risk of chronic health conditions via persistent oxidative stress. In this work, two kinds of acute exposure experiments of iron oxide (Fe2O3 and Fe3O4) nanoparticles in cells and in vivo were conducted to evaluate the GSH levels and GST activity. Our current research presented Fe3O4 nanoparticles at lower concentrations (≤100 μg/ml) seem to be more toxic to the human bronchial epithelial cells as their consumption of GSH and decrease of GST activity. The catalysis activity of Fe3O4 nanoparticles per se may contribute to the intracellular GSH consumption along with inhibition of glutathione-S-transferase class mu 1 and P (GSTM1 and GSTP1) active site and expression decrease of GSTM1 and GSTP1. Accordingly, the GSH consumption and decrease in GST activity directed to the further lipid peroxidation regarded as an earlier marker for toxicity evaluation of iron oxide nanoparticles, and relevant intervention may be effective for prevention of respiratory exposure induced damage from iron oxide nanoparticles.
Collapse
Affiliation(s)
- Wanjun Zhang
- Departmental of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Jinling Gao
- Departmental of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China; Department of Infection Management Service, Dushu Lake Hospital Affiliated of Soochow University, Suzhou 215000, China
| | - Lin Lu
- Departmental of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Tsendmaa Bold
- Departmental of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Xin Li
- Departmental of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Shuo Wang
- Departmental of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Zhishang Chang
- Departmental of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Jing Chen
- Departmental of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Xiao Kong
- Departmental of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Yuxin Zheng
- Departmental of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Mingliang Zhang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 300384, China.
| | - Jinglong Tang
- Departmental of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
13
|
Liu H, Zhang H, Cheng D, Tan K, Ye T, Ma H, Li S, Zheng H. Differential responses of a pi-class glutathione S-transferase (CnGSTp) expression and antioxidant status between golden and brown noble scallops under pathogenic stress. FISH & SHELLFISH IMMUNOLOGY 2020; 105:144-151. [PMID: 32652299 DOI: 10.1016/j.fsi.2020.07.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/30/2020] [Accepted: 07/02/2020] [Indexed: 06/11/2023]
Abstract
Glutathione S-transferases (GSTs) play important roles in immunity by protecting organisms against the damage of reactive oxygen species (ROS). In this study, a pi-class GST cDNA sequence was first cloned from noble scallop Chlamys nobilis (named CnGSTp). The full length cDNA of CnGSTp was 922 bp, encoding a cytosolic protein of 202 amino acids residues, with predicted molecular masses of 23.1 kDa. Then an acute Vibrio Parahaemolyticus challenge experiment was conducted by using the Golden and Brown noble scallops with different total carotenoids content (TCC), and CnGSTp expression level, TCC and ROS level was separately determined. The results showed that ROS and CnGSTp expression levels were significantly up-regulate under Vibrio Parahaemolyticus challenge than the control group (P < 0.05). The Golden scallops showed significantly higher CnGSTp expression level and lower ROS level in hemocytes than the Brown ones (P < 0.05). Moreover, there is a significantly positive correlation between TCC and ROS in the Golden scallops. The present results revealed that CnGSTp plays important roles in immune response and carotenoids play assistant roles in antioxidant defense system under pathogenic stress in the noble scallop.
Collapse
Affiliation(s)
- Hongxing Liu
- Key Laboratory of Marine Biotechnology of Guangdong Province, Shantou University, Shantou, 515063, China; Mariculture Research Center for Subtropical Shellfish & Algae of Guangdong Province, Shantou 515063, China; STU-UMT Joint Shellfish Research Laboratory, Shantou University, Shantou 515063, China
| | - Hongkuan Zhang
- Key Laboratory of Marine Biotechnology of Guangdong Province, Shantou University, Shantou, 515063, China; Mariculture Research Center for Subtropical Shellfish & Algae of Guangdong Province, Shantou 515063, China; STU-UMT Joint Shellfish Research Laboratory, Shantou University, Shantou 515063, China
| | - Dewei Cheng
- Key Laboratory of Marine Biotechnology of Guangdong Province, Shantou University, Shantou, 515063, China; Mariculture Research Center for Subtropical Shellfish & Algae of Guangdong Province, Shantou 515063, China; STU-UMT Joint Shellfish Research Laboratory, Shantou University, Shantou 515063, China
| | - Karsoon Tan
- Key Laboratory of Marine Biotechnology of Guangdong Province, Shantou University, Shantou, 515063, China; Mariculture Research Center for Subtropical Shellfish & Algae of Guangdong Province, Shantou 515063, China; STU-UMT Joint Shellfish Research Laboratory, Shantou University, Shantou 515063, China
| | - Ting Ye
- Key Laboratory of Marine Biotechnology of Guangdong Province, Shantou University, Shantou, 515063, China; Mariculture Research Center for Subtropical Shellfish & Algae of Guangdong Province, Shantou 515063, China; STU-UMT Joint Shellfish Research Laboratory, Shantou University, Shantou 515063, China
| | - Hongyu Ma
- Key Laboratory of Marine Biotechnology of Guangdong Province, Shantou University, Shantou, 515063, China; Mariculture Research Center for Subtropical Shellfish & Algae of Guangdong Province, Shantou 515063, China; STU-UMT Joint Shellfish Research Laboratory, Shantou University, Shantou 515063, China
| | - Shengkang Li
- Key Laboratory of Marine Biotechnology of Guangdong Province, Shantou University, Shantou, 515063, China; Mariculture Research Center for Subtropical Shellfish & Algae of Guangdong Province, Shantou 515063, China; STU-UMT Joint Shellfish Research Laboratory, Shantou University, Shantou 515063, China
| | - Huaiping Zheng
- Key Laboratory of Marine Biotechnology of Guangdong Province, Shantou University, Shantou, 515063, China; Mariculture Research Center for Subtropical Shellfish & Algae of Guangdong Province, Shantou 515063, China; STU-UMT Joint Shellfish Research Laboratory, Shantou University, Shantou 515063, China.
| |
Collapse
|
14
|
Jin L, Jagatheesan G, Lynch J, Guo L, Conklin DJ. Crotonaldehyde-induced vascular relaxation and toxicity: Role of endothelium and transient receptor potential ankyrin-1 (TRPA1). Toxicol Appl Pharmacol 2020; 398:115012. [PMID: 32320793 PMCID: PMC7375699 DOI: 10.1016/j.taap.2020.115012] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 04/13/2020] [Accepted: 04/15/2020] [Indexed: 01/25/2023]
Abstract
INTRODUCTION Crotonaldehyde (CR) is an electrophilic α,β-unsaturated aldehyde present in foods and beverages and is a minor metabolite of 1,3-butadiene. CR is a product of incomplete combustion, and is at high levels in smoke of cigarettes and structural fires. Exposure to CR has been linked to cardiopulmonary toxicity and cardiovascular disease. OBJECTIVE The purpose of this study was to examine the direct effects of CR in murine blood vessels (aorta and superior mesenteric artery, SMA) using an in vitro system. METHODS AND RESULTS CR induced concentration-dependent (1-300 μM) relaxations (75-80%) in phenylephrine (PE) precontracted aorta and SMA. Because the SMA was 20× more sensitive to CR than aorta (SMA EC50 3.8 ± 0.5 μM; aorta EC50 76.0 ± 2.0 μM), mechanisms of CR relaxation were studied in SMA. The CR-induced relaxation at low concentrations (1-30 μM) was inhibited by: 1) mechanically-impaired endothelium; 2) Nω-Nitro-L-arginine methyl ester hydrochloride (L-NAME); 3) guanylyl cyclase (GC) inhibitor (ODQ); 4) transient receptor potential ankyrin-1 (TRPA1) antagonist (A967079); and, 5) by non-vasoactive level of nicotine (1 μM). Similarly, a TRPA1 agonist, allyl isothiocyanate (AITC; mustard oil), stimulated SMA relaxation dependent on TRPA1, endothelium, NO, and GC. Consistent with these mechanisms, TRPA1 was present in the SMA endothelium. CR, at higher concentrations (100-300 μM), induced tension oscillations (spasms) and irreversibly impaired contractility (a vasotoxic effect enhanced by impaired endothelium). CONCLUSIONS CR relaxation depends on a functional endothelium and TRPA1, whereas vasotoxicity is enhanced by endothelium dysfunction. Thus, CR is both vasoactive and vasotoxic along a concentration continuum.
Collapse
Affiliation(s)
- L Jin
- Department of Anesthesiology, Critical Care and Pain Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China; Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, KY, USA; Christina Lee Brown Envirome Institute, University of Louisville, Louisville, KY, USA; Diabetes and Obesity Center, University of Louisville, Louisville, KY, USA; American Heart Association-Tobacco Regulation Center, University of Louisville, Louisville, KY, USA
| | - G Jagatheesan
- Christina Lee Brown Envirome Institute, University of Louisville, Louisville, KY, USA; Diabetes and Obesity Center, University of Louisville, Louisville, KY, USA; American Heart Association-Tobacco Regulation Center, University of Louisville, Louisville, KY, USA
| | - J Lynch
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, KY, USA; Christina Lee Brown Envirome Institute, University of Louisville, Louisville, KY, USA; Diabetes and Obesity Center, University of Louisville, Louisville, KY, USA; American Heart Association-Tobacco Regulation Center, University of Louisville, Louisville, KY, USA
| | - L Guo
- Christina Lee Brown Envirome Institute, University of Louisville, Louisville, KY, USA; Diabetes and Obesity Center, University of Louisville, Louisville, KY, USA; American Heart Association-Tobacco Regulation Center, University of Louisville, Louisville, KY, USA
| | - D J Conklin
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, KY, USA; Christina Lee Brown Envirome Institute, University of Louisville, Louisville, KY, USA; Diabetes and Obesity Center, University of Louisville, Louisville, KY, USA; American Heart Association-Tobacco Regulation Center, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
15
|
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
|
16
|
The Role of Antioxidants in Ameliorating Cyclophosphamide-Induced Cardiotoxicity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:4965171. [PMID: 32454939 PMCID: PMC7238386 DOI: 10.1155/2020/4965171] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 04/09/2020] [Accepted: 04/21/2020] [Indexed: 12/11/2022]
Abstract
The chemotherapeutic and immunosuppressive agent cyclophosphamide has previously been shown to induce complications within the setting of bone marrow transplantation. More recently, cardiotoxicity has been shown to be a dose-limiting factor during cyclophosphamide therapy, and cardiooncology is getting wider attention. Though mechanism of cyclophosphamide-induced cardiotoxicity is not completely understood, it is thought to encompass oxidative and nitrative stress. As such, this review focuses on antioxidants and their role in preventing or ameliorating cyclophosphamide-induced cardiotoxicity. It will give special emphasis to the cardioprotective effects of natural, plant-derived antioxidants that have garnered significant interest in recent times.
Collapse
|
17
|
Zhang Y, Xue W, Zhang W, Yuan Y, Zhu X, Wang Q, Wei Y, Yang D, Yang C, Chen Y, Sun Y, Wang S, Huang K, Zheng L. Histone methyltransferase G9a protects against acute liver injury through GSTP1. Cell Death Differ 2019; 27:1243-1258. [PMID: 31515511 PMCID: PMC7206029 DOI: 10.1038/s41418-019-0412-8] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 08/01/2019] [Accepted: 08/16/2019] [Indexed: 11/24/2022] Open
Abstract
Acute liver injury is commonly caused by bacterial endotoxin/lipopolysaccharide (LPS), and by drug overdose such as acetaminophen (APAP). The exact role of epigenetic modification in acute liver injury remains elusive. Here, we investigated the role of histone methyltransferase G9a in LPS- or APAP overdose-induced acute liver injury. Under d-galactosamine sensitization, liver-specific G9a-deficient mice (L-G9a−/−) exhibited 100% mortality after LPS injection, while the control and L-G9a+/− littermates showed very mild mortality. Moreover, abrogation of hepatic G9a or inhibiting the methyltransferase activity of G9a aggravated LPS-induced liver damage. Similarly, under sublethal APAP overdose, L-G9a−/− mice displayed more severe liver injury. Mechanistically, ablation of G9a inhibited H3K9me1 levels at the promoters of Gstp1/2, two liver detoxifying enzymes, and consequently suppressed their transcription. Notably, treating L-G9a−/− mice with recombinant mouse GSTP1 reversed the LPS- or APAP overdose-induced liver damage. Taken together, we identify a novel beneficial role of G9a-GSTP1 axis in protecting against acute liver injury.
Collapse
Affiliation(s)
- Yu Zhang
- Tongji School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, PR China
| | - Weili Xue
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei, PR China
| | - Wenquan Zhang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei, PR China
| | - Yangmian Yuan
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei, PR China
| | - Xiuqin Zhu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei, PR China
| | - Qing Wang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei, PR China
| | - Yujuan Wei
- Tongji School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, PR China
| | - Dong Yang
- Tongji School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, PR China
| | - Chen Yang
- Tongji School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, PR China
| | - Yan Chen
- Tongji School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, PR China
| | - Yu Sun
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei, PR China
| | - Shun Wang
- Department of Blood Transfusion, Wuhan Hospital of Traditional and Western Medicine, Wuhan, 430022, PR China
| | - Kun Huang
- Tongji School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, PR China.
| | - Ling Zheng
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei, PR China.
| |
Collapse
|
18
|
Conklin DJ, Guo Y, Nystoriak MA, Jagatheesan G, Obal D, Kilfoil PJ, Hoetker JD, Guo L, Bolli R, Bhatnagar A. TRPA1 channel contributes to myocardial ischemia-reperfusion injury. Am J Physiol Heart Circ Physiol 2019; 316:H889-H899. [PMID: 30735434 PMCID: PMC6483018 DOI: 10.1152/ajpheart.00106.2018] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 01/18/2019] [Accepted: 01/31/2019] [Indexed: 11/22/2022]
Abstract
Myocardial ischemia-reperfusion (I/R) results in the generation of free radicals, accumulation of lipid peroxidation-derived unsaturated aldehydes, variable angina (pain), and infarction. The transient receptor potential ankyrin 1 (TRPA1) mediates pain signaling and is activated by unsaturated aldehydes, including acrolein and 4-hydroxynonenal. The contribution of TRPA1 (a Ca2+-permeable channel) to I/R-induced myocardial injury is unknown. We tested the hypothesis that cardiac TRPA1 confers myocyte sensitivity to aldehyde accumulation and promotes I/R injury. Although basal cardiovascular function in TRPA1-null mice was similar to that in wild-type (WT) mice, infarct size was significantly smaller in TRPA1-null mice than in WT mice (34.1 ± 9.3 vs. 14.3 ± 9.9% of the risk region, n = 8 and 7, respectively, P < 0.05), despite a similar I/R-induced area at risk (40.3 ±8.4% and 42.2 ± 11.3% for WT and TRPA1-null mice, respectively) after myocardial I/R (30 min of ischemia followed by 24 h of reperfusion) in situ. Positive TRPA1 immunofluorescence was present in murine and human hearts and was colocalized with connexin43 at intercalated disks in isolated murine cardiomyocytes. Cardiomyocyte TRPA1 was confirmed by quantitative RT-PCR, DNA sequencing, Western blot analysis, and electrophysiology. A role of TRPA1 in cardiomyocyte toxicity was demonstrated in isolated cardiomyocytes exposed to acrolein, an I/R-associated toxin that induces Ca2+ accumulation and hypercontraction, effects significantly blunted by HC-030031, a TRPA1 antagonist. Protection induced by HC-030031 was quantitatively equivalent to that induced by SN-6, a Na+/Ca2+ exchange inhibitor, further supporting a role of Ca2+ overload in acrolein-induced cardiomyocyte toxicity. These data indicate that cardiac TRPA1 activation likely contributes to I/R injury and, thus, that TRPA1 may be a novel therapeutic target for decreasing myocardial I/R injury. NEW & NOTEWORTHY Transient receptor potential ankyrin 1 (TRPA1) activation mediates increased blood flow, edema, and pain reception, yet its role in myocardial ischemia-reperfusion (I/R) injury is unknown. Genetic ablation of TRPA1 significantly decreased myocardial infarction after I/R in mice. Functional TRPA1 in cardiomyocytes was enriched in intercalated disks and contributed to acrolein-induced Ca2+ overload and hypercontraction. These data indicate that I/R activation of TRPA1 worsens myocardial infarction; TRPA1 may be a potential target to mitigate I/R injury.
Collapse
Affiliation(s)
- Daniel J Conklin
- Diabetes and Obesity Center, University of Louisville , Louisville, Kentucky
- Envirome Institute, University of Louisville , Louisville, Kentucky
- Department of Medicine, University of Louisville , Louisville, Kentucky
| | - Yiru Guo
- Institute of Molecular Cardiology, University of Louisville , Louisville, Kentucky
- Department of Medicine, University of Louisville , Louisville, Kentucky
| | - Matthew A Nystoriak
- Diabetes and Obesity Center, University of Louisville , Louisville, Kentucky
- Envirome Institute, University of Louisville , Louisville, Kentucky
- Department of Medicine, University of Louisville , Louisville, Kentucky
| | - Ganapathy Jagatheesan
- Diabetes and Obesity Center, University of Louisville , Louisville, Kentucky
- Envirome Institute, University of Louisville , Louisville, Kentucky
- Department of Medicine, University of Louisville , Louisville, Kentucky
| | - Detlef Obal
- Diabetes and Obesity Center, University of Louisville , Louisville, Kentucky
- Institute of Molecular Cardiology, University of Louisville , Louisville, Kentucky
- Department of Anesthesiology and Perioperative Medicine, University of Louisville , Louisville, Kentucky
| | - Peter J Kilfoil
- Smidt Heart Institute, Cedars-Sinai Hospital , Los Angeles, California
| | - Joseph David Hoetker
- Diabetes and Obesity Center, University of Louisville , Louisville, Kentucky
- Envirome Institute, University of Louisville , Louisville, Kentucky
- Department of Medicine, University of Louisville , Louisville, Kentucky
| | - Luping Guo
- Diabetes and Obesity Center, University of Louisville , Louisville, Kentucky
- Envirome Institute, University of Louisville , Louisville, Kentucky
- Department of Medicine, University of Louisville , Louisville, Kentucky
| | - Roberto Bolli
- Diabetes and Obesity Center, University of Louisville , Louisville, Kentucky
- Institute of Molecular Cardiology, University of Louisville , Louisville, Kentucky
- Department of Medicine, University of Louisville , Louisville, Kentucky
| | - Aruni Bhatnagar
- Diabetes and Obesity Center, University of Louisville , Louisville, Kentucky
- Envirome Institute, University of Louisville , Louisville, Kentucky
- Department of Medicine, University of Louisville , Louisville, Kentucky
| |
Collapse
|
19
|
Totzeck M, Schuler M, Stuschke M, Heusch G, Rassaf T. Cardio-oncology - strategies for management of cancer-therapy related cardiovascular disease. Int J Cardiol 2019; 280:163-175. [PMID: 30661849 DOI: 10.1016/j.ijcard.2019.01.038] [Citation(s) in RCA: 133] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 12/15/2018] [Accepted: 01/10/2019] [Indexed: 02/07/2023]
Abstract
Current therapy of advanced cancers is based on several modalities including radiotherapy, cytotoxic chemotherapy, molecularly targeted inhibitors and antibodies targeting immune checkpoints. All of those these modalities can negatively impact the cardiovascular system, and there is considerable experience in relation to radiotherapy and chemotherapy. In contrast, the knowledge base on cardiovascular toxicities of novel agents targeting signal transduction pathways and immune regulation is quite limited. In particular, potential late effects are of concern as cardiovascular pathology can negatively impact quality of life and prognosis in cancer survivors, particularly when additional cardiovascular risk factors are present. Treatment-associated adverse events include hypertension, venous thromboembolism, coronary artery disease, valvular heart disease, heart failure and arrhythmias. Early diagnosis of subclinical cardiotoxic effects of cancer therapies remains challenging. Integrated care, as provided by multidisciplinary cardio-oncology teams is the best option for prevention, diagnosis and treatment of cardiovascular diseases associated with cancer therapy. This review considers the cardiotoxic effects of specific cancer therapies and discusses novel diagnostic and therapeutic approaches as a reference for optimizing the care of cancer patients receiving novel cancer therapies.
Collapse
Affiliation(s)
- Matthias Totzeck
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, Medical Faculty, University Hospital Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Martin Schuler
- Department of Medical Oncology, West German Cancer Center, Medical Faculty, University Hospital Essen, Hufelandstr. 55, 45147 Essen, Germany; German Cancer Consortium (DKTK), Partner site University Hospital Essen, Hufelandstrasse 55, 45147 Essen, Germany
| | - Martin Stuschke
- Department of Radiation Oncology, West German Cancer Center, Medical Faculty, University Hospital Essen, Hufelandstr. 55, 45147 Essen, Germany; German Cancer Consortium (DKTK), Partner site University Hospital Essen, Hufelandstrasse 55, 45147 Essen, Germany
| | - Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, Medical Faculty, University Hospital Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Tienush Rassaf
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, Medical Faculty, University Hospital Essen, Hufelandstr. 55, 45147 Essen, Germany.
| |
Collapse
|
20
|
Oleaga C, Riu A, Rothemund S, Lavado A, McAleer CW, Long CJ, Persaud K, Narasimhan NS, Tran M, Roles J, Carmona-Moran CA, Sasserath T, Elbrecht DH, Kumanchik L, Bridges LR, Martin C, Schnepper MT, Ekman G, Jackson M, Wang YI, Note R, Langer J, Teissier S, Hickman JJ. Investigation of the effect of hepatic metabolism on off-target cardiotoxicity in a multi-organ human-on-a-chip system. Biomaterials 2018; 182:176-190. [PMID: 30130706 DOI: 10.1016/j.biomaterials.2018.07.062] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 07/31/2018] [Indexed: 12/30/2022]
Abstract
Regulation of cosmetic testing and poor predictivity of preclinical drug studies has spurred efforts to develop new methods for systemic toxicity. Current in vitro assays do not fully represent physiology, often lacking xenobiotic metabolism. Functional human multi-organ systems containing iPSC derived cardiomyocytes and primary hepatocytes were maintained under flow using a low-volume pumpless system in a serum-free medium. The functional readouts for contractile force and electrical conductivity enabled the non-invasive study of cardiac function. The presence of the hepatocytes in the system induced cardiotoxic effects from cyclophosphamide and reduced them for terfenadine due to drug metabolism, as expected from each compound's pharmacology. A computational fluid dynamics simulation enabled the prediction of terfenadine-fexofenadine pharmacokinetics, which was validated by HPLC-MS. This in vitro platform recapitulates primary aspects of the in vivo crosstalk between heart and liver and enables pharmacological studies, involving both organs in a single in vitro platform. The system enables non-invasive readouts of cardiotoxicity of drugs and their metabolites. Hepatotoxicity can also be evaluated by biomarker analysis and change in metabolic function. Integration of metabolic function in toxicology models can improve adverse effects prediction in preclinical studies and this system could also be used for chronic studies as well.
Collapse
Affiliation(s)
- Carlota Oleaga
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32826, USA
| | - Anne Riu
- L'Oreal Research, and Innovation Division, Aulnay-sous-Bois, France
| | - Sandra Rothemund
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32826, USA
| | - Andrea Lavado
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32826, USA
| | - Christopher W McAleer
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32826, USA; Hesperos, Inc., 3259 Progress Dr, Room 158, Orlando, FL 32826, USA
| | - Christopher J Long
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32826, USA; Hesperos, Inc., 3259 Progress Dr, Room 158, Orlando, FL 32826, USA
| | - Keisha Persaud
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32826, USA
| | | | - My Tran
- Hesperos, Inc., 3259 Progress Dr, Room 158, Orlando, FL 32826, USA
| | - Jeffry Roles
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32826, USA
| | - Carlos A Carmona-Moran
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32826, USA
| | - Trevor Sasserath
- Hesperos, Inc., 3259 Progress Dr, Room 158, Orlando, FL 32826, USA
| | - Daniel H Elbrecht
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32826, USA; Hesperos, Inc., 3259 Progress Dr, Room 158, Orlando, FL 32826, USA
| | - Lee Kumanchik
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32826, USA
| | | | - Candace Martin
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32826, USA
| | - Mark T Schnepper
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32826, USA
| | - Gail Ekman
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32826, USA
| | - Max Jackson
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32826, USA
| | - Ying I Wang
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32826, USA; Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Reine Note
- L'Oreal Research, and Innovation Division, Aulnay-sous-Bois, France
| | - Jessica Langer
- L'Oreal Research, and Innovation Division, Clark, NJ, USA
| | - Silvia Teissier
- L'Oreal Research, and Innovation Division, Aulnay-sous-Bois, France
| | - James J Hickman
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32826, USA.
| |
Collapse
|
21
|
Liu W, Zhai X, Wang W, Zheng B, Zhang Z, Fan X, Chen Y, Wang J. Aldehyde dehydrogenase 2 activation ameliorates cyclophosphamide-induced acute cardiotoxicity via detoxification of toxic aldehydes and suppression of cardiac cell death. J Mol Cell Cardiol 2018; 121:134-144. [PMID: 29981795 DOI: 10.1016/j.yjmcc.2018.07.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 06/03/2018] [Accepted: 07/04/2018] [Indexed: 01/27/2023]
Abstract
Cyclophosphamide (CY)-induced acute cardiotoxicity is a common side effect which is dose dependent. It is reported that up to 20% of patients received high dose of CY treatment suffered from acute cardiac dysfunction. However, the effective intervention strategies and related mechanisms are still largely unknown. We aimed to investigate the effects of aldehyde dehydrogenase 2 (ALDH2), an important endogenous cardioprotective enzyme, on CY-induced acute cardiotoxicity and the underlying mechanisms. It was found that ALDH2 knockout (KO) mice were more sensitive to CY-induced acute cardiotoxicity, presenting as higher serum levels of creatine kinase-MB isoform and lactate dehydrogenase, and significantly reduced myocardial contractility compared with C57BL/6 (WT) mice. In addition, cardiac cell death, especially necrosis, was obviously increased in ALDH2 KO mice compared with WT mice after CY treatment. Furthermore, accumulation of toxic aldehydes such as acrolein and 4-HNE and reactive oxygen species (ROS) in the myocardium were significantly elevated after CY in ALDH2 KO mice. Importantly, ALDH2 activation by Alda-1 pretreatment markedly attenuated CY-induced accumulation of toxic aldehydes, cardiac cell death and cardiac dysfunction, without affecting CY's anti-tumor efficacy. In conclusion, the cardioprotective effects of ALDH2 activation against CY-induced acute cardiotoxicity are exerted via reducing toxic aldehydes accumulation and potentially interrupting the acrolein-ROS-aldehydes vicious circles, and thus alleviates myocardial cell death, without antagonizing the anti-tumor efficacy of CY. Therefore, ALDH2 might be a promising prevention and treatment target for CY-induced acute cardiotoxicity.
Collapse
Affiliation(s)
- Wenwen Liu
- Department of Emergency Medicine, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China; Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaoxuan Zhai
- Department of Emergency Medicine, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China; Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Wenjun Wang
- Department of Emergency Medicine, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China; Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Boyuan Zheng
- Department of Emergency Medicine, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China; Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Zhenxiao Zhang
- Department of Emergency Medicine, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China; Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China; Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Xinhui Fan
- Department of Emergency Medicine, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China; Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Yuguo Chen
- Department of Emergency Medicine, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China; Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China.
| | - Jiali Wang
- Department of Emergency Medicine, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China; Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China.
| |
Collapse
|
22
|
Baba SP, Zhang D, Singh M, Dassanayaka S, Xie Z, Jagatheesan G, Zhao J, Schmidtke VK, Brittian KR, Merchant ML, Conklin DJ, Jones SP, Bhatnagar A. Deficiency of aldose reductase exacerbates early pressure overload-induced cardiac dysfunction and autophagy in mice. J Mol Cell Cardiol 2018; 118:183-192. [PMID: 29627295 PMCID: PMC6205513 DOI: 10.1016/j.yjmcc.2018.04.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 03/29/2018] [Accepted: 04/03/2018] [Indexed: 12/21/2022]
Abstract
Pathological cardiac hypertrophy is associated with the accumulation of lipid peroxidation-derived aldehydes such as 4-hydroxy-trans-2-nonenal (HNE) and acrolein in the heart. These aldehydes are metabolized via several pathways, of which aldose reductase (AR) represents a broad-specificity route for their elimination. We tested the hypothesis that by preventing aldehyde removal, AR deficiency accentuates the pathological effects of transverse aortic constriction (TAC). We found that the levels of AR in the heart were increased in mice subjected to TAC for 2 weeks. In comparison with wild-type (WT), AR-null mice showed lower ejection fraction, which was exacerbated 2 weeks after TAC. Levels of atrial natriuretic peptide and myosin heavy chain were higher in AR-null than in WT TAC hearts. Deficiency of AR decreased urinary levels of the acrolein metabolite, 3-hydroxypropylmercapturic acid. Deletion of AR did not affect the levels of the other aldehyde-metabolizing enzyme - aldehyde dehydrogenase 2 in the heart, or its urinary product - (N-Acetyl-S-(2-carboxyethyl)-l-cystiene). AR-null hearts subjected to TAC showed increased accumulation of HNE- and acrolein-modified proteins, as well as increased AMPK phosphorylation and autophagy. Superfusion with HNE led to a greater increase in p62, LC3II formation, and GFP-LC3-II punctae formation in AR-null than WT cardiac myocytes. Pharmacological inactivation of JNK decreased HNE-induced autophagy in AR-null cardiac myocytes. Collectively, these results suggest that during hypertrophy the accumulation of lipid peroxidation derived aldehydes promotes pathological remodeling via excessive autophagy, and that metabolic detoxification of these aldehydes by AR may be essential for maintaining cardiac function during early stages of pressure overload.
Collapse
Affiliation(s)
- Shahid P Baba
- Diabetes and Obesity Center, University of Louisville, Louisville, KY, United States.
| | - Deqing Zhang
- Diabetes and Obesity Center, University of Louisville, Louisville, KY, United States
| | - Mahavir Singh
- Department of Physiology, University of Louisville, Louisville, KY, United States
| | - Sujith Dassanayaka
- Diabetes and Obesity Center, University of Louisville, Louisville, KY, United States
| | - Zhengzhi Xie
- Diabetes and Obesity Center, University of Louisville, Louisville, KY, United States
| | - Ganapathy Jagatheesan
- Diabetes and Obesity Center, University of Louisville, Louisville, KY, United States
| | - Jingjing Zhao
- Diabetes and Obesity Center, University of Louisville, Louisville, KY, United States
| | - Virginia K Schmidtke
- Diabetes and Obesity Center, University of Louisville, Louisville, KY, United States
| | - Kenneth R Brittian
- Diabetes and Obesity Center, University of Louisville, Louisville, KY, United States
| | - Michael L Merchant
- Divisions of Nephrology and Hypertension and the Institute of Molecular Cardiology, University of Louisville, Louisville, KY, United States
| | - Daniel J Conklin
- Diabetes and Obesity Center, University of Louisville, Louisville, KY, United States
| | - Steven P Jones
- Diabetes and Obesity Center, University of Louisville, Louisville, KY, United States
| | - Aruni Bhatnagar
- Diabetes and Obesity Center, University of Louisville, Louisville, KY, United States
| |
Collapse
|
23
|
Coyle JP, Rinaldi RJ, Johnson GT, Bourgeois MM, McCluskey JD, Harbison RD. Reduced oxygen tension culturing conditionally alters toxicogenic response of differentiated H9c2 cardiomyoblasts to acrolein. Toxicol Mech Methods 2018; 28:488-498. [PMID: 29564938 DOI: 10.1080/15376516.2018.1455785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Acrolein is a reactive electrophilic aldehyde known to cause mitochondrial dysfunction, oxidative stress, and dysregulation of signaling transduction in vitro. Most in vitro systems employ standard cell culture maintenance conditions of 95% air/5% CO2, translating to a culture oxygen tension of approximately 20%, far above most physiological tissues. The purpose of this investigation was to examine whether low-serum, retinoic acid differentiated H9c2 cells were less sensitive to acrolein insult when cultured under reduced oxygen tension. H9c2 cells were maintained separately in 20% and 5% oxygen, differentiated for 5 d, and then exposed to acrolein for 30 min in media containing varying concentrations of tricarboxylic acid and glycolytic substrates, followed by fresh medium replacement. Cells were then assessed for MTT reduction at 2 h and 24 h after acrolein insult. We showed that pyruvate supplementation in combination with lowered oxygen culturing significantly attenuated acrolein-induced viability loss at 24 h. Poly(ADP-ribose) polymerase inhibition and EGTA preferentially provided partial rescue to low oxygen cultures, but not for standard cultures. Collectively, these results offer evidence supporting altered toxicogenic response of H9c2 during physiologically relevant oxygen tension culturing.
Collapse
Affiliation(s)
- Jayme P Coyle
- a Department of Environmental and Occupational Heath , College of Public Health, University of South Florida , Tampa , FL , USA
| | - Robert J Rinaldi
- b Department of Integrative Biology , College of Arts and Sciences, University of South Florida , Tampa , FL , USA
| | - Giffe T Johnson
- a Department of Environmental and Occupational Heath , College of Public Health, University of South Florida , Tampa , FL , USA
| | - Marie M Bourgeois
- a Department of Environmental and Occupational Heath , College of Public Health, University of South Florida , Tampa , FL , USA
| | - James D McCluskey
- a Department of Environmental and Occupational Heath , College of Public Health, University of South Florida , Tampa , FL , USA
| | - Raymond D Harbison
- a Department of Environmental and Occupational Heath , College of Public Health, University of South Florida , Tampa , FL , USA
| |
Collapse
|
24
|
The Positive Effects of Exercise in Chemotherapy-Related Cardiomyopathy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1000:103-129. [DOI: 10.1007/978-981-10-4304-8_8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
25
|
Conklin DJ, Malovichko MV, Zeller I, Das TP, Krivokhizhina TV, Lynch BH, Lorkiewicz P, Agarwal A, Wickramasinghe N, Haberzettl P, Sithu SD, Shah J, O’Toole TE, Rai SN, Bhatnagar A, Srivastava S. Biomarkers of Chronic Acrolein Inhalation Exposure in Mice: Implications for Tobacco Product-Induced Toxicity. Toxicol Sci 2017; 158:263-274. [PMID: 28482051 PMCID: PMC5837482 DOI: 10.1093/toxsci/kfx095] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Exposure to tobacco smoke, which contains several harmful and potentially harmful constituents such as acrolein increases cardiovascular disease (CVD) risk. Although high acrolein levels induce pervasive cardiovascular injury, the effects of low-level exposure remain unknown and sensitive biomarkers of acrolein toxicity have not been identified. Identification of such biomarkers is essential to assess the toxicity of acrolein present at low levels in the ambient air or in new tobacco products such as e-cigarettes. Hence, we examined the systemic effects of chronic (12 weeks) acrolein exposure at concentrations similar to those found in tobacco smoke (0.5 or 1 ppm). Acrolein exposure in mice led to a 2- to 3-fold increase in its urinary metabolite 3-hydroxypropyl mercapturic acid (3-HPMA) with an attendant increase in pulmonary levels of the acrolein-metabolizing enzymes, glutathione S-transferase P and aldose reductase, as well as several Nrf2-regulated antioxidant proteins. Markers of pulmonary endoplasmic reticulum stress and inflammation were unchanged. Exposure to acrolein suppressed circulating levels of endothelial progenitor cells (EPCs) and specific leukocyte subsets (eg, GR-1+ cells, CD19+ B-cells, CD4+ T-cells; CD11b+ monocytes) whilst other subsets (eg, CD8+ cells, NK1.1+ cells, Ly6C+ monocytes) were unchanged. Chronic acrolein exposure did not affect systemic glucose tolerance, platelet-leukocyte aggregates or microparticles in blood. These findings suggest that circulating levels of EPCs and specific leukocyte populations are sensitive biomarkers of inhaled acrolein injury and that low-level (<0.5 ppm) acrolein exposure (eg, in secondhand smoke, vehicle exhaust, e-cigarettes) could increase CVD risk by diminishing endothelium repair or by suppressing immune cells or both.
Collapse
Affiliation(s)
- Daniel J. Conklin
- American Heart Association – Tobacco Regulation and Addiction Center
- Diabetes and Obesity Center
- Institute of Molecular Cardiology
- Division of Cardiovascular Medicine, Department of Medicine School of Medicine
| | - Marina V. Malovichko
- American Heart Association – Tobacco Regulation and Addiction Center
- Diabetes and Obesity Center
- Institute of Molecular Cardiology
- Division of Cardiovascular Medicine, Department of Medicine School of Medicine
| | - Iris Zeller
- American Heart Association – Tobacco Regulation and Addiction Center
- Diabetes and Obesity Center
- Institute of Molecular Cardiology
- Division of Cardiovascular Medicine, Department of Medicine School of Medicine
| | - Trinath P. Das
- American Heart Association – Tobacco Regulation and Addiction Center
- Diabetes and Obesity Center
- Institute of Molecular Cardiology
- Division of Cardiovascular Medicine, Department of Medicine School of Medicine
| | - Tatiana V. Krivokhizhina
- American Heart Association – Tobacco Regulation and Addiction Center
- Diabetes and Obesity Center
- Institute of Molecular Cardiology
- Division of Cardiovascular Medicine, Department of Medicine School of Medicine
| | - Blake H. Lynch
- American Heart Association – Tobacco Regulation and Addiction Center
- Diabetes and Obesity Center
- Institute of Molecular Cardiology
- Division of Cardiovascular Medicine, Department of Medicine School of Medicine
| | - Pawel Lorkiewicz
- American Heart Association – Tobacco Regulation and Addiction Center
- Diabetes and Obesity Center
- Institute of Molecular Cardiology
- Division of Cardiovascular Medicine, Department of Medicine School of Medicine
| | - Abhinav Agarwal
- American Heart Association – Tobacco Regulation and Addiction Center
- Diabetes and Obesity Center
- Institute of Molecular Cardiology
- Division of Cardiovascular Medicine, Department of Medicine School of Medicine
| | - Nalinie Wickramasinghe
- American Heart Association – Tobacco Regulation and Addiction Center
- Diabetes and Obesity Center
- Institute of Molecular Cardiology
- Division of Cardiovascular Medicine, Department of Medicine School of Medicine
| | - Petra Haberzettl
- American Heart Association – Tobacco Regulation and Addiction Center
- Diabetes and Obesity Center
- Institute of Molecular Cardiology
- Division of Cardiovascular Medicine, Department of Medicine School of Medicine
| | - Srinivas D. Sithu
- American Heart Association – Tobacco Regulation and Addiction Center
- Diabetes and Obesity Center
| | - Jasmit Shah
- American Heart Association – Tobacco Regulation and Addiction Center
- Diabetes and Obesity Center
- School of Public Health & Information Sciences, University of Louisville, Louisville, Kentucky 40202
| | - Timothy E. O’Toole
- American Heart Association – Tobacco Regulation and Addiction Center
- Diabetes and Obesity Center
- Institute of Molecular Cardiology
- Division of Cardiovascular Medicine, Department of Medicine School of Medicine
| | - Shesh N. Rai
- American Heart Association – Tobacco Regulation and Addiction Center
- Diabetes and Obesity Center
- School of Public Health & Information Sciences, University of Louisville, Louisville, Kentucky 40202
| | - Aruni Bhatnagar
- American Heart Association – Tobacco Regulation and Addiction Center
- Diabetes and Obesity Center
- Institute of Molecular Cardiology
- Division of Cardiovascular Medicine, Department of Medicine School of Medicine
| | - Sanjay Srivastava
- American Heart Association – Tobacco Regulation and Addiction Center
- Diabetes and Obesity Center
- Institute of Molecular Cardiology
- Division of Cardiovascular Medicine, Department of Medicine School of Medicine
| |
Collapse
|
26
|
Abstract
Actin-binding proteins are proteins that could bind to actin or actin fibers. As a member of actin-binding proteins, Transgelin-2 is expressed in smooth muscle cells and non-smooth muscle cells, and its gene, TAGLN2, is differently expressed in all cells and tissues. The deregulation of Transgelin-2 is considered to be correlated with progression of many kinds of diseases, especially the development of malignant tumors, such as invasion, metastasis, and resistance, yet the function and mechanism of action of Transgelin-2 remain elusive. Therefore, we reviewed the basic characteristics and function of Transgelin-2 and its biological role in various types of diseases in order to provide the theoretical basis for further research and new perspectives on cancer development.
Collapse
Affiliation(s)
- Ti Meng
- Department of Pharmacy, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, China
| | - Leichao Liu
- Department of Pharmacy, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, China
| | - Ruifang Hao
- Department of Pharmacy, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, China
| | - Siying Chen
- Department of Pharmacy, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, China
| | - Yalin Dong
- Department of Pharmacy, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
27
|
El-Agamy DS, Elkablawy MA, Abo-Haded HM. Modulation of cyclophosphamide-induced cardiotoxicity by methyl palmitate. Cancer Chemother Pharmacol 2017; 79:399-409. [DOI: 10.1007/s00280-016-3233-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 12/26/2016] [Indexed: 11/29/2022]
|
28
|
Burcham PC. Acrolein and Human Disease: Untangling the Knotty Exposure Scenarios Accompanying Several Diverse Disorders. Chem Res Toxicol 2016; 30:145-161. [DOI: 10.1021/acs.chemrestox.6b00310] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Philip C. Burcham
- Pharmacology, Pharmacy & Anaesthesiology Unit, School of Medicine and Pharmacology, The University of Western Australia, Crawley, Western Australia 6007, Australia
| |
Collapse
|
29
|
Conklin DJ, Haberzettl P, Jagatheesan G, Kong M, Hoyle GW. Role of TRPA1 in acute cardiopulmonary toxicity of inhaled acrolein. Toxicol Appl Pharmacol 2016; 324:61-72. [PMID: 27592100 DOI: 10.1016/j.taap.2016.08.028] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 08/25/2016] [Accepted: 08/29/2016] [Indexed: 12/25/2022]
Abstract
Acrolein is a highly toxic, volatile, unsaturated aldehyde generated during incomplete combustion as in tobacco smoke and indoor fires. Because the transient receptor potential ankyrin 1 (TRPA1) channel mediates tobacco smoke-induced lung injury, we assessed its role in high-level acrolein-induced toxicity in mice. Acrolein (100-275ppm, 10-30min) caused upper airway epithelial sloughing, bradypnea and oral gasping, hypothermia, cardiac depression and mortality. Male wild-type mice (WT, C57BL/6; 5-52weeks) were significantly more sensitive to high-level acrolein than age-matched, female WT mice. Both male and female TRPA1-null mice were more sensitive to acrolein-induced mortality than age- and sex-matched WT mice. Acrolein exposure increased lung weight:body weight ratios and lung albumin and decreased plasma albumin to a greater extent in TRPA1-null than in WT mice. Lung and plasma protein-acrolein adducts were not increased in acrolein-exposed TRPA1-null mice compared with WT mice. To assess TRPA1-dependent protective mechanisms, respiratory parameters were monitored by telemetry. TRPA1-null mice had a slower onset of breathing rate suppression ('respiratory braking') than WT mice suggesting TRPA1 mediates this protective response. Surprisingly, WT male mice treated either with a TRPA1 antagonist (HC030031; 200mg/kg) alone or with combined TRPA1 (100mg/kg) and TRPV1 (capsazepine, 10mg/kg) antagonists at 30min post-acrolein exposure (i.e., "real world" delay in treatment) were significantly protected from acrolein-induced mortality. These data show TRPA1 protects against high-level acrolein-induced toxicity in a sex-dependent manner. Post-exposure TRPA1 antagonism also protected against acrolein-induced mortality attesting to a complex role of TRPA1 in cardiopulmonary injury.
Collapse
Affiliation(s)
- Daniel J Conklin
- Diabetes and Obesity Center, Institute of Molecular Cardiology, Division of Cardiovascular Medicine, Department of Medicine, School of Medicine, University of Louisville, Louisville, KY 40292, United States.
| | - Petra Haberzettl
- Diabetes and Obesity Center, Institute of Molecular Cardiology, Division of Cardiovascular Medicine, Department of Medicine, School of Medicine, University of Louisville, Louisville, KY 40292, United States
| | - Ganapathy Jagatheesan
- Diabetes and Obesity Center, Institute of Molecular Cardiology, Division of Cardiovascular Medicine, Department of Medicine, School of Medicine, University of Louisville, Louisville, KY 40292, United States
| | - Maiying Kong
- Department of Bioinformatics and Biostatistics, School of Public Health & Information Sciences, University of Louisville, Louisville, KY 40292, United States
| | - Gary W Hoyle
- Department of Environmental and Occupational Health Sciences, School of Public Health & Information Sciences, University of Louisville, Louisville, KY 40292, United States
| |
Collapse
|
30
|
Conklin DJ. Acute cardiopulmonary toxicity of inhaled aldehydes: role of TRPA1. Ann N Y Acad Sci 2016; 1374:59-67. [PMID: 27152448 DOI: 10.1111/nyas.13055] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Revised: 02/26/2016] [Accepted: 03/07/2016] [Indexed: 12/13/2022]
Abstract
Inhalation of high-level volatile aldehydes, as present in smoke from wildfires and in tobacco smoke, is associated with both acute and chronic cardiopulmonary morbidity and mortality, but the underlying mechanisms are unclear. The transient receptor potential ankyrin 1 (TRPA1) protein forms a cation channel (irritant receptor) that mediates tobacco smoke-induced airway and lung injury, yet the role of TRPA1 in the cardiovascular toxicity of aldehyde exposure is unclear. Physiologically, airway-located TRPA1 activation triggers an irritant response (e.g., coughing and "respiratory braking") that alters the rate and depth of breathing to reduce exposure. Acrolein (2-propenal), a volatile, unsaturated aldehyde, activates TRPA1. Acrolein was used as a chemical weapon in World War I and is present at high levels in wildfires and tobacco smoke. Acrolein is thought to contribute to pulmonary and cardiovascular injury caused by tobacco smoke exposure, although the role of TRPA1 in cardiovascular toxicity is unclear. This minireview addresses this gap in our knowledge by exploring literature and recent data indicating a connection between TRPA1 and cardiovascular as well as pulmonary injury due to inhaled aldehydes.
Collapse
Affiliation(s)
- Daniel J Conklin
- Diabetes and Obesity Center, Institute of Molecular Cardiology, Division of Cardiovascular Medicine, Department of Medicine, University of Louisville, Louisville, Kentucky
| |
Collapse
|
31
|
Madeddu C, Deidda M, Piras A, Cadeddu C, Demurtas L, Puzzoni M, Piscopo G, Scartozzi M, Mercuro G. Pathophysiology of cardiotoxicity induced by nonanthracycline chemotherapy. J Cardiovasc Med (Hagerstown) 2016; 17 Suppl 1:e12-e18. [PMID: 27183520 DOI: 10.2459/jcm.0000000000000376] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The risk and mechanism of chemotherapy-induced cardiotoxicity (CTX) vary depending on the type and intensity of the anticancer regimen. Myriad chemotherapeutic drugs produce adverse cardiovascular effects such as arterial hypertension, heart failure, and thromboembolic events. Among the numerous classes of these drugs, anthracyclines have been studied most extensively because of their overt cardiovascular effects and the high associated incidence of heart failure. However, CTX might also be caused by other types of chemotherapeutic agents, including alkylating agents (cyclophosphamide, ifosfamide), platinum agents, antimetabolites (5-fluorouracil, capecitabine), antibiotics (mitoxantrone, mitomycin, bleomycin), and antimicrotubule agents (taxanes). Here, we review the incidence, clinical impact, and potential mechanisms of CTX associated with nonanthracycline chemotherapy used for cancer patients. The published data support a marked increase in CTX risk, particularly with certain drugs such as 5-fluorouracil and cisplatin. Each anticancer regimen is associated with distinct modes of heart damage, both symptomatic and asymptomatic. However, the underlying mechanisms of CTX have been established only in a few cases, and only few nonanthracycline chemotherapeutics (mitoxantrone, mitomycin, ifosfamide) act through a recognizable mechanism and show a predictable dose dependence. Lastly, nonanthracycline chemotherapy can induce both chronic lesions, such as systolic dysfunction, and acute lesions, such as the ischemia that occurs within hours or days after treatment. An increased understanding of the incidence, mechanisms, and potential therapeutic targets of CTX induced by various nonanthracycline chemotherapeutic agents is clearly required.
Collapse
Affiliation(s)
- Clelia Madeddu
- aDepartment of Medical Sciences Mario Aresu, Unit of Medical Oncology bDepartment of Medical Sciences Mario Aresu, Unit of Cardiology and Angiology, University Hospital Cagliari, University of Cagliari, Cagliari cDivision of Cardiology, Istituto Nazionale per lo Studio e la Cura dei Tumori 'Fondazione Giovanni Pascale' - IRCCS, Naples, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Song Y, Zhang C, Wang C, Zhao L, Wang Z, Dai Z, Lin S, Kang H, Ma X. Ferulic Acid against Cyclophosphamide-Induced Heart Toxicity in Mice by Inhibiting NF-κB Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2016; 2016:1261270. [PMID: 26881001 PMCID: PMC4736310 DOI: 10.1155/2016/1261270] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 09/17/2015] [Indexed: 02/05/2023]
Abstract
The purpose of the present study was to elucidate the protective effects of ferulic acid (FA) against cyclophosphamide- (CTX-) induced changes in mice. Forty-eight male ICR mice were divided into four groups. Control group was intraperitoneally (i.p.) injected with 200 μL of phosphate buffer saline (PBS). Model group was intraperitoneally injected with a single dose of CTX (200 mg/kg). FA (50 mg/kg) and FA (100 mg/kg) groups were intraperitoneally injected with a single dose of CTX (200 mg/kg) followed by the intragastric treatment with FA (50, 100 mg/kg) for 7 consecutive days. After 12 d, the mice were sacrificed to analyze the hematological, biochemical, histological parameters and mechanism research. The results indicated that FA significantly decreased the serum levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), creatine kinase (CK), lactate dehydrogenase (LDH), interleukin-6 (IL-6), IL-1β, and tumor necrosis factor-α (TNF-α) in CTX-injected mice. In addition, FA effectively reduced the total numbers of white blood cells (WBCs), red blood cells, platelets, and hemoglobin content. FA also obviously attenuated the histological changes of the heart tissues caused by CTX. Moreover, Western blot demonstrated that FA inhibited the phosphorylations of NF-κB signaling pathway in CTX-stimulated cardiac tissues. In conclusion, FA might be considered as an effective agent in the amelioration of the heart toxicity resulting from CTX treatment.
Collapse
Affiliation(s)
- Yafan Song
- Department of Cardiology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an 710004, China
| | - Chunyan Zhang
- Department of Cardiology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an 710004, China
| | - Congxia Wang
- Department of Cardiology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an 710004, China
| | - Ling Zhao
- Department of Cardiology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an 710004, China
| | - Zheng Wang
- Department of Oncology, The Central Hospital of Xi'an, Xi'an 710003, China
| | - Zhijun Dai
- Department of Oncology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an 710004, China
| | - Shuai Lin
- Department of Oncology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an 710004, China
| | - Huafeng Kang
- Department of Oncology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an 710004, China
| | - Xiaobin Ma
- Department of Oncology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an 710004, China
| |
Collapse
|
33
|
Li Y, Deng H, Ju L, Zhang X, Zhang Z, Yang Z, Wang L, Hou Z, Zhang Y. Screening and validation for plasma biomarkers of nephrotoxicity based on metabolomics in male rats. Toxicol Res (Camb) 2016; 5:259-267. [PMID: 30090342 PMCID: PMC6062367 DOI: 10.1039/c5tx00171d] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 11/02/2015] [Indexed: 12/16/2022] Open
Abstract
Currently, drug-induced nephrotoxicity is widespread and seriously affects human health. However, the conventional indexes of renal function lack sensitivity, leading to a delay in the detection of nephrotoxicity. Therefore, we need to identify more sensitive indexes for evaluating nephrotoxicity. In this study, we used gentamicin (100 mg kg-1), etimicin (100 mg kg-1) and amphotericin B (4 mg kg-1) to establish renal injury models in rats, and we collected information using ultra-performance liquid chromatography quadrupole time-of-flight mass spectrometry in the screening stage. Thirteen nephrotoxicity metabolites were selected after multivariate statistical and integration analyses. Then, we conducted trend analysis to select 5 nephrotoxicity biomarkers [thymidine, LysoPC(16:1), LysoPC(18:4), LysoPC(20:5), and LysoPC(22:5)] whose content changed consistently at different timepoints after drug administration. To verify the sensitivity and specificity of these biomarkers for nephrotoxicity, receiver operating characteristic (ROC) and support vector machine (SVM) analyses were applied. The area under the curve of the 5 biomarkers were 0.806-0.901 at the 95% confidence interval according to the ROC analysis. We used the SVM classified model to verify these biomarkers, and the prediction rate was 95.83%. Therefore, the 5 biomarkers have strong sensitivity and high accuracy; these biomarkers are more sensitive indexes for evaluating renal function to identify nephrotoxicity and initiate prompt treatment.
Collapse
Affiliation(s)
- Yubo Li
- Tianjin State Key Laboratory of Modern Chinese Medicine , School of Traditional Chinese Materia Medica , Tianjin University of Traditional Chinese Medicine , 312 Anshan west Road , Tianjin 300193 , China
| | - Haoyue Deng
- Tianjin State Key Laboratory of Modern Chinese Medicine , School of Traditional Chinese Materia Medica , Tianjin University of Traditional Chinese Medicine , 312 Anshan west Road , Tianjin 300193 , China
| | - Liang Ju
- Tianjin State Key Laboratory of Modern Chinese Medicine , School of Traditional Chinese Materia Medica , Tianjin University of Traditional Chinese Medicine , 312 Anshan west Road , Tianjin 300193 , China
| | - Xiuxiu Zhang
- Tianjin State Key Laboratory of Modern Chinese Medicine , School of Traditional Chinese Materia Medica , Tianjin University of Traditional Chinese Medicine , 312 Anshan west Road , Tianjin 300193 , China
| | - Zhenzhu Zhang
- Tianjin State Key Laboratory of Modern Chinese Medicine , School of Traditional Chinese Materia Medica , Tianjin University of Traditional Chinese Medicine , 312 Anshan west Road , Tianjin 300193 , China
| | - Zhen Yang
- Tianjin State Key Laboratory of Modern Chinese Medicine , School of Traditional Chinese Materia Medica , Tianjin University of Traditional Chinese Medicine , 312 Anshan west Road , Tianjin 300193 , China
| | - Lei Wang
- Tianjin State Key Laboratory of Modern Chinese Medicine , School of Traditional Chinese Materia Medica , Tianjin University of Traditional Chinese Medicine , 312 Anshan west Road , Tianjin 300193 , China
| | - Zhiguo Hou
- Tianjin State Key Laboratory of Modern Chinese Medicine , School of Traditional Chinese Materia Medica , Tianjin University of Traditional Chinese Medicine , 312 Anshan west Road , Tianjin 300193 , China
| | - Yanjun Zhang
- Tianjin State Key Laboratory of Modern Chinese Medicine , Tianjin University of Traditional Chinese Medicine , 312 Anshan west Road , Tianjin 300193 , China . ; ; Tel: +86-22-59596221
| |
Collapse
|
34
|
Ginseng alleviates cyclophosphamide-induced hepatotoxicity via reversing disordered homeostasis of glutathione and bile acid. Sci Rep 2015; 5:17536. [PMID: 26625948 PMCID: PMC4667192 DOI: 10.1038/srep17536] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 11/02/2015] [Indexed: 12/19/2022] Open
Abstract
Cyclophosphamide (CP), a chemotherapeutic agent, is restricted due to its side effects, especially hepatotoxicity. Ginseng has often been clinically used with CP in China, but whether and how ginseng reduces the hepatotoxicity is unknown. In this study, the hepatoprotective effects and mechanisms under the combined usage were investigated. It was found that ginseng could ameliorate CP-induced elevations of ALP, ALT, ALS, MDA and hepatic deterioration, enhance antioxidant enzymes’ activities and GSH’s level. Metabolomics study revealed that 33 endogenous metabolites were changed by CP, 19 of which were reversed when ginseng was co-administrated via two main pathways, i.e., GSH metabolism and primary bile acids synthesis. Furthermore, ginseng could induce expression of GCLC, GCLM, GS and GST, which associate with the disposition of GSH, and expression of FXR, CYP7A1, NTCP and MRP 3, which play important roles in the synthesis and transport of bile acids. In addition, NRF 2, one of regulatory elements on the expression of GCLC, GCLM, GS, GST, NTCP and MRP3, was up-regulated when ginseng was co-administrated. In conclusion, ginseng could alleviate CP-induced hepatotoxicity via modulating the disordered homeostasis of GSH and bile acid, which might be mediated by inducing the expression of NRF 2 in liver.
Collapse
|
35
|
McGarry DJ, Chakravarty P, Wolf CR, Henderson CJ. Altered protein S-glutathionylation identifies a potential mechanism of resistance to acetaminophen-induced hepatotoxicity. J Pharmacol Exp Ther 2015; 355:137-44. [PMID: 26311813 PMCID: PMC4631951 DOI: 10.1124/jpet.115.227389] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 08/25/2015] [Indexed: 01/01/2023] Open
Abstract
Acetaminophen (APAP) is the most commonly used over-the-counter analgesic. However, hepatotoxicity induced by APAP is a major clinical issue, and the factors that define sensitivity to APAP remain unclear. We have previously demonstrated that mice nulled for glutathione S-transferase Pi (GSTP) are resistant to APAP-induced hepatotoxicity. This study aims to exploit this difference to delineate pathways of importance in APAP toxicity. We used mice nulled for GSTP and heme oxygenase-1 oxidative stress reporter mice, together with a novel nanoflow liquid chromatography-tandem mass spectrometry methodology to investigate the role of oxidative stress, cell signaling, and protein S-glutathionylation in APAP hepatotoxicity. We provide evidence that the sensitivity difference between wild-type and Gstp1/2(-/-) mice is unrelated to the ability of APAP to induce oxidative stress, despite observing significant increases in c-Jun N-terminal kinase and extracellular signal-regulated kinase phosphorylation in wild-type mice. The major difference in response to APAP was in the levels of protein S-glutathionylation: Gstp1/2(-/-) mice exhibited a significant increase in the number of S-glutathionylated proteins compared with wild-type animals. Remarkably, these S-glutathionylated proteins are involved in oxidative phosphorylation, respiratory complexes, drug metabolism, and mitochondrial apoptosis. Furthermore, we found that S-glutathionylation of the rate-limiting glutathione-synthesizing enzyme, glutamate cysteine ligase, was markedly increased in Gstp1/2(-/-) mice in response to APAP. The data demonstrate that S-glutathionylation provides an adaptive response to APAP and, as a consequence, suggest that this is an important determinant in APAP hepatotoxicity. This work identifies potential novel avenues associated with cell survival for the treatment of chemical-induced hepatotoxicity.
Collapse
Affiliation(s)
- David J McGarry
- Molecular Pharmacology Group, School of Medicine, Jacqui Wood Cancer Centre, University of Dundee, Dundee, United Kingdom (D.J.M., C.R.W., C.J.H.); and Bioinformatics and Biostatistics Group, Cancer Research UK London Research Institute, London, United Kingdom (P.C.)
| | - Probir Chakravarty
- Molecular Pharmacology Group, School of Medicine, Jacqui Wood Cancer Centre, University of Dundee, Dundee, United Kingdom (D.J.M., C.R.W., C.J.H.); and Bioinformatics and Biostatistics Group, Cancer Research UK London Research Institute, London, United Kingdom (P.C.)
| | - C Roland Wolf
- Molecular Pharmacology Group, School of Medicine, Jacqui Wood Cancer Centre, University of Dundee, Dundee, United Kingdom (D.J.M., C.R.W., C.J.H.); and Bioinformatics and Biostatistics Group, Cancer Research UK London Research Institute, London, United Kingdom (P.C.)
| | - Colin J Henderson
- Molecular Pharmacology Group, School of Medicine, Jacqui Wood Cancer Centre, University of Dundee, Dundee, United Kingdom (D.J.M., C.R.W., C.J.H.); and Bioinformatics and Biostatistics Group, Cancer Research UK London Research Institute, London, United Kingdom (P.C.)
| |
Collapse
|