1
|
Xu C, Wu M, Yu W, Xie D, Wang Q, Chen B, Xi Y, Yu L, Yan Y, Yamamoto T, Koyama H, Zhao H, Cheng J. High Uric Acid Orchestrates Ferroptosis to Promote Cardiomyopathy Via ROS-GPX4 Signaling. Antioxid Redox Signal 2024; 41:1134-1149. [PMID: 39113539 DOI: 10.1089/ars.2023.0473] [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: 08/29/2024]
Abstract
Aims: High uric acid (HUA), as a pro-oxidant, plays a significant role in the pathophysiology of cardiovascular disease. Studies have indicated that elevated uric acid levels can adversely affect cardiovascular health. Nevertheless, the impact of hyperuricemia on cardiomyopathy remains uncertain. Further research is needed to elucidate the relationship between HUA and cardiomyopathy, shedding light on its potential implications for heart health. Results: We demonstrated that uricase knockout (Uox-KO) mice accelerated the development of cardiomyopathy, causing significantly impaired cardiac function and myocardial fibrosis. Meanwhile, the mitochondrial morphology was destroyed, the lipid peroxidation products increased in number and the antioxidant function was weakened. In addition, we evaluated the effects of ferrostatin-1 (Fer-1), the ferroptosis inhibitor. Myocardial damage can be reversed by the Fer-1 treatment caused by HUA combined with doxorubicin (DOX) treatment. Benzbromarone, a uric acid-lowering drug, decreases myocardial fibrosis, and ferroptosis by alleviating hyperuricemia in Uox-KO mice by DOX administration. In vitro, we observed that the activity of cardiomyocytes treated with HUA combined with DOX decreased significantly, and lipid reactive oxygen species (ROS) increased significantly. Afterward, we demonstrated that HUA can promote oxidative stress in DOX, characterized by increased mitochondrial ROS, and downregulate protein levels of glutathione peroxidase 4 (GPX4). N-acetyl-l-cysteine, an antioxidant, inhibits the process by which HUA promotes DOX-induced ferroptosis by increasing the GPX4 expression. Innovation: We verified that HUA can exacerbate myocardial damage. This has clinical implications for the treatment of cardiac damage in patients with hyperuricemia. Conclusions: Our data suggested that HUA promotes the cardiomyopathy. HUA promotes DOX-induced ferroptosis by increasing oxidative stress and downregulating GPX4. Antioxid. Redox Signal. 41, 1134-1149.
Collapse
Affiliation(s)
- Chenxi Xu
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, China
- Institute of Metabolism and Cell Death, Helmholtz Munich, 85764 Neuherberg, Bavaria, Germany
| | - Mengni Wu
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Wei Yu
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - De Xie
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Qiang Wang
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Binyang Chen
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Yuemei Xi
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Linqian Yu
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Yunbo Yan
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Tetsuya Yamamoto
- Department of Diabetes, Endocrinology, and Clinical Immunology, Hyogo College of Medicine, Nishinomiya, Japan
| | - Hidenori Koyama
- Department of Diabetes, Endocrinology, and Clinical Immunology, Hyogo College of Medicine, Nishinomiya, Japan
| | - Hong Zhao
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Translational Medicine for Nucleic Acid Metabolism and Regulation, Xiamen, China
| | - Jidong Cheng
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, China
- Department of Diabetes, Endocrinology, and Clinical Immunology, Hyogo College of Medicine, Nishinomiya, Japan
- Xiamen Key Laboratory of Translational Medicine for Nucleic Acid Metabolism and Regulation, Xiamen, China
| |
Collapse
|
2
|
Wan H, Yang X, Zhang Y, Liu X, Li Y, Qin Y, Yan H, Gui L, Li K, Zhang L, Yang L, Zhang B, Wang Y. Polyphenol-Reinforced Glycocalyx-Like Hydrogel Coating Induced Myocardial Regeneration and Immunomodulation. ACS NANO 2024; 18:21512-21522. [PMID: 39096486 DOI: 10.1021/acsnano.4c06332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/05/2024]
Abstract
Although minimally invasive interventional occluders can effectively seal heart defect tissue, they still have some limitations, including poor endothelial healing, intense inflammatory response, and thrombosis formation. Herein, a polyphenol-reinforced medicine/peptide glycocalyx-like coating was prepared on cardiac occluders. A coating consisting of carboxylated chitosan, epigallocatechin-3-gallate (EGCG), tanshinone IIA sulfonic sodium (TSS), and hyaluronic acid grafted with 3-aminophenylboronic acid was prepared. Subsequently, the mercaptopropionic acid-GGGGG-Arg-Glu-Asp-Val peptide was grafted by the thiol-ene "click" reaction. The coating showed good hydrophilicity and free radical-scavenging ability and could release EGCG-TSS. The results of biological experiments suggested that the coating could reduce thrombosis by promoting endothelialization, and promote myocardial repair by regulating the inflammatory response. The functions of regulating cardiomyocyte apoptosis and metabolism were confirmed, and the inflammatory regulatory functions of the coating were mainly dependent on the NF-kappa B and TNF signaling pathway.
Collapse
Affiliation(s)
- Huining Wan
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Xiaohui Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Yutong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Xiyu Liu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Yanyan Li
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Yumei Qin
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Hui Yan
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Lan Gui
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Ke Li
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Longjian Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Li Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Bo Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
- Bioengineering Department, University of California, Los Angeles, California 90095, United States
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
| |
Collapse
|
3
|
Zhang LL, Chen GH, Tang RJ, Xiong YY, Pan Q, Jiang WY, Gong ZT, Chen C, Li XS, Yang YJ. Levosimendan Reverses Cardiac Malfunction and Cardiomyocyte Ferroptosis During Heart Failure with Preserved Ejection Fraction via Connexin 43 Signaling Activation. Cardiovasc Drugs Ther 2024; 38:705-718. [PMID: 36881213 DOI: 10.1007/s10557-023-07441-4] [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] [Accepted: 02/13/2023] [Indexed: 03/08/2023]
Abstract
PURPOSE In recent decades, the occurrence of heart failure with preserved ejection fraction (HFpEF) has outweighed that of heart failure with reduced ejection fraction by degrees, but few drugs have been demonstrated to improve long-term clinical outcomes in patients with HFpEF. Levosimendan, a calcium-sensitizing cardiotonic agent, improves decompensated heart failure clinically. However, the anti-HFpEF activities of levosimendan and underlying molecular mechanisms are unclear. METHODS In this study, a double-hit HFpEF C57BL/6N mouse model was established, and levosimendan (3 mg/kg/week) was administered to HFpEF mice aged 13 to 17 weeks. Different biological experimental techniques were used to verify the protective effects of levosimendan against HFpEF. RESULTS After four weeks of drug treatment, left ventricular diastolic dysfunction, cardiac hypertrophy, pulmonary congestion, and exercise exhaustion were significantly alleviated. Junction proteins in the endothelial barrier and between cardiomyocytes were also improved by levosimendan. Among the gap junction channel proteins, connexin 43, which was especially highly expressed in cardiomyocytes, mediated mitochondrial protection. Furthermore, levosimendan reversed mitochondrial malfunction in HFpEF mice, as evidenced by increased mitofilin and decreased ROS, superoxide anion, NOX4, and cytochrome C levels. Interestingly, after levosimendan administration, myocardial tissue from HFpEF mice showed restricted ferroptosis, indicated by an increased GSH/GSSG ratio; upregulated GPX4, xCT, and FSP-1 expression; and reduced intracellular ferrous ion, MDA, and 4-HNE levels. CONCLUSION Regular long-term levosimendan administration can benefit cardiac function in a mouse model of HFpEF with metabolic syndromes (namely, obesity and hypertension) by activating connexin 43-mediated mitochondrial protection and sequential ferroptosis inhibition in cardiomyocytes.
Collapse
MESH Headings
- Animals
- Ferroptosis/drug effects
- Heart Failure/drug therapy
- Heart Failure/physiopathology
- Heart Failure/metabolism
- Simendan/pharmacology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Connexin 43/metabolism
- Mice, Inbred C57BL
- Disease Models, Animal
- Stroke Volume/drug effects
- Ventricular Function, Left/drug effects
- Male
- Signal Transduction/drug effects
- Mice
- Mitochondria, Heart/drug effects
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/pathology
- Ventricular Dysfunction, Left/drug therapy
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Dysfunction, Left/metabolism
- Cardiotonic Agents/pharmacology
Collapse
Affiliation(s)
- Li-Li Zhang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100037, China
| | - Gui-Hao Chen
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100037, China
| | - Rui-Jie Tang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Yu-Yan Xiong
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qi Pan
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100037, China
| | - Wen-Yang Jiang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100037, China
| | - Zhao-Ting Gong
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100037, China
| | - Cheng Chen
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100037, China
| | - Xiao-Song Li
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100037, China
| | - Yue-Jin Yang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100037, China.
| |
Collapse
|
4
|
Efentakis P, Varela A, Lamprou S, Papanagnou ED, Chatzistefanou M, Christodoulou A, Davos CH, Gavriatopoulou M, Trougakos I, Dimopoulos MA, Terpos E, Andreadou I. Implications and hidden toxicity of cardiometabolic syndrome and early-stage heart failure in carfilzomib-induced cardiotoxicity. Br J Pharmacol 2024; 181:2964-2990. [PMID: 38679957 DOI: 10.1111/bph.16391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 07/25/2023] [Accepted: 09/12/2023] [Indexed: 05/01/2024] Open
Abstract
BACKGROUND AND PURPOSE Cancer therapy-related cardiovascular adverse events (CAEs) in presence of comorbidities, are in the spotlight of the cardio-oncology guidelines. Carfilzomib (Cfz), indicated for relapsed/refractory multiple myeloma (MM), presents with serious CAEs. MM is often accompanied with co-existing comorbidities. However, Cfz use in MM patients with cardiometabolic syndrome (CMS) or in heart failure with reduced ejection fraction (HFrEF), is questionable. EXPERIMENTAL APPROACH ApoE-/- and C57BL6/J male mice received 14 weeks Western Diet (WD) (CMS models). C57BL6/J male mice underwent permanent LAD ligation for 14 days (early-stage HFrEF model). CMS- and HFrEF-burdened mice received Cfz for two consecutive or six alternate days. Daily metformin and atorvastatin administrations were performed additionally to Cfz, as prophylactic interventions. Mice underwent echocardiography, while proteasome activity, biochemical and molecular analyses were conducted. KEY RESULTS CMS did not exacerbate Cfz left ventricular (LV) dysfunction, whereas Cfz led to metabolic complications in both CMS models. Cfz induced autophagy and Ca2+ homeostasis dysregulation, whereas metformin and atorvastatin prevented Cfz-mediated LV dysfunction and molecular deficits in the CMS-burdened myocardium. Early-stage HFrEF led to depressed LV function and increased protein phosphatase 2A (PP2A) activity. Cfz further increased myocardial PP2A activity, inflammation and Ca2+-cycling dysregulation. Metformin co-administration exerted an anti-inflammatory potential on the myocardium without improving LV function. CONCLUSION AND IMPLICATIONS CMS and HFrEF seem to exacerbate Cfz-induced CAEs, by presenting metabolism-related hidden toxicity and PP2A-related cardiac inflammation, respectively. Metformin retains its prophylactic potential in the presence of CMS, while mitigating inflammation and Ca2+ signalling dysregulation in the HFrEF myocardium.
Collapse
Affiliation(s)
- Panagiotis Efentakis
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Aimilia Varela
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Sofia Lamprou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Michail Chatzistefanou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Andriana Christodoulou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Maria Gavriatopoulou
- Department of Clinical Therapeutics, Plasma Cell Dyscrasias Unit, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Ioannis Trougakos
- Department of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Meletios Athanasios Dimopoulos
- Department of Clinical Therapeutics, Plasma Cell Dyscrasias Unit, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Evangelos Terpos
- Department of Clinical Therapeutics, Plasma Cell Dyscrasias Unit, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
5
|
Schelz Z, Muddather HF, Jaski FS, Bózsity N, Zupkó I. An In Vitro Investigation of the Antiproliferative and Antimetastatic Effects of Levosimendan: Potential Drug Repurposing for Cervical Cancer. Curr Issues Mol Biol 2024; 46:6566-6579. [PMID: 39057033 PMCID: PMC11275392 DOI: 10.3390/cimb46070391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/20/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024] Open
Abstract
Cervical cancer presents a significant challenge to the global health of women. Despite substantial advances in human papillomavirus (HPV)-related cervical cancer vaccines, non-HPV-related cervical cancer is still waiting novel therapeutic options. Drug repurposing has provided a promising approach to improve cancer therapy in recent years. Our study aimed to explore the potential in vitro antineoplastic effects of levosimendan on cervical cancer cells. The antiproliferative effects of levosimendan were investigated on cervical cancer cells using a standard MTT assay. Fluorescent double staining was performed to identify its ability to induce apoptosis and necrosis. The possible mechanism of action of levosimendan was explored using cell-cycle analysis. Furthermore, antimetastatic effects were investigated using a wound-healing assay and a Boyden chamber assay. Our results revealed that levosimendan exhibited the highest growth-inhibitory effect in the HPV-negative C33A cell line. However, the effects were modest compared to the standard agent, cisplatin. Cell-cycle analysis detected that levosimendan can induce cell-cycle arrest in C33A cells by increasing the G1 and G2/M phases, decreasing the S phase, and enhancing the hypodiploid subG1 population. Levosimendan inhibited cell migration and invasion in a concentration-dependent manner. As levosimendan showed antimetastatic efficacy, it could be considered for repurposing to contribute to overcoming resistance to therapy in cervical cancer.
Collapse
Affiliation(s)
| | | | | | | | - István Zupkó
- Institute of Pharmacodynamics and Biopharmacy, Faculty of Pharmacy, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary; (Z.S.); (H.F.M.); (F.S.J.); (N.B.)
| |
Collapse
|
6
|
Zhou N, Wei S, Sun T, Xie S, Liu J, Li W, Zhang B. Recent progress in the role of endogenous metal ions in doxorubicin-induced cardiotoxicity. Front Pharmacol 2023; 14:1292088. [PMID: 38143497 PMCID: PMC10748411 DOI: 10.3389/fphar.2023.1292088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 11/28/2023] [Indexed: 12/26/2023] Open
Abstract
Doxorubicin is a widely used anticancer drug in clinical practice for the treatment of various human tumors. However, its administration is associated with cardiotoxicity. Administration of doxorubicin with low side effects for cancer treatment and prevention are, accordingly, urgently required. The human body harbors various endogenous metal ions that exert substantial influences. Consequently, extensive research has been conducted over several decades to investigate the potential of targeting endogenous metal ions to mitigate doxorubicin's side effects and impede tumor progression. In recent years, there has been a growing body of research indicating the potential efficacy of metal ion-associated therapeutic strategies in inhibiting doxorubicin-induced cardiotoxicity (DIC). These strategies offer a combination of favorable safety profiles and potential clinical utility. Alterations in intracellular levels of metal ions have been found to either facilitate or mitigate the development of DIC. For instance, ferroptosis, a cellular death mechanism, and metal ions such as copper, zinc, and calcium have been identified as significant contributors to DIC. This understanding can contribute to advancements in cancer treatment and provide valuable insights for mitigating the cardiotoxic effects of other therapeutic drugs. Furthermore, potential therapeutic strategies have been investigated to alleviate DIC in clinical settings. The ultimate goal is to improve the efficacy and safety of Dox and offer valuable insights for future research in this field.
Collapse
Affiliation(s)
- Ni Zhou
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Clinical Pharmacy, Central South University, Changsha, Hunan, China
- School of Pharmacy, Central South University, Changsha, Hunan, China
| | - Shanshan Wei
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Clinical Pharmacy, Central South University, Changsha, Hunan, China
| | - Taoli Sun
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Suifen Xie
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Clinical Pharmacy, Central South University, Changsha, Hunan, China
- School of Pharmacy, Central South University, Changsha, Hunan, China
| | - Jian Liu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Clinical Pharmacy, Central South University, Changsha, Hunan, China
| | - Wenqun Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Clinical Pharmacy, Central South University, Changsha, Hunan, China
| | - Bikui Zhang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Clinical Pharmacy, Central South University, Changsha, Hunan, China
- School of Pharmacy, Central South University, Changsha, Hunan, China
| |
Collapse
|
7
|
李 新, 闫 爱, 常 晋, 李 汾, 朱 娟. [Hesperetin Alleviates Doxorubicin-Induced Cytotoxicity in H9c2 Cells by Activating SIRT1/NRF2 Signaling]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2023; 54:947-953. [PMID: 37866951 PMCID: PMC10579077 DOI: 10.12182/20230960207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Indexed: 10/24/2023]
Abstract
Objective To investigate whether hesperetin (Hes) alleviates doxorubicin (DOX)-induced cardiomyocytotoxicity by reducing oxidative stress via regulating silent information regulator 1 (SIRT1)/nuclear transcription factor E2-related factor 2 (NRF2) signaling in H9c2 cells. Methods H9c2 cells were treated with DOX to establish the cardiotoxicity model and were randomly assigned to four groups, a control group (Control) and three treatment groups, receiving respectively DOX (the DOX group), Hes+DOX (the DOX+Hes group), and Hes+SIRT1 inhibitor EX527+DOX (the DOX+Hes+EX527 group). Cellular morphology was observed by the light microscope. Cell viability was evaluated by CCK-8. DOX-induced apoptosis in H9c2 cells was examined by flow cytometry. The levels of reactive oxygen species (ROS) in the H9c2 cells of the four groups were determied with 2'-7'-dichlorodihydrofluorescein diacetate (DCFH-DA) staining. The activities of lactate dehydrogenase (LDH), superoxide dismutase (SOD), catalase (CAT), and SIRT1 as well as the malondialdehyde (MDA) content were measured using ELISA kits. The expressions of cleaved caspase-3, cytochrome c, SIRT1, Ac-FOXO1, NRF2, and heme oxygenase 1 (HO-1) were determined by Western blot. Results Compared with the Control group, the DOX group showed swollen cellular morphology, decreased cell density and viability, and increased LDH activity in the medium ( P<0.01); both apoptosis and the expression of cleaved caspase-3 and cytochrome c increased ( P<0.01); the activities of CAT and SOD decreased while the contents of MDA and ROS increased ( P<0.01); the expression of SIRT1, NRF2, and HO-1 decreased, the activity of SIRT1 decreased, and the expression of Ac-FOXO1 increased ( P<0.01). Compared with the DOX group, the DOX+Hes group showed improved cellular morphology, increased cell density and viability, and decreased LDH activity in the medium ( P<0.01); the apoptosis and the expression of cleaved caspase-3 and cytochrome c decreased ( P<0.01); the activities of CAT and SOD increased while the levels of MDA and ROS decreased ( P<0.01); the expression of SIRT1, NRF2, and HO-1 increased, the activity of SIRT1 increased, and the expression of Ac-FOXO1 decreased ( P<0.01). Comparison of the findings for the DOX+Hes group and the DOX+Hes+EX527 group showed that EX527 could block the protective effects of Hes against DOX-induced cell injury, oxidative stress, and SIRT1/NRF2 signaling. Conclusion Hes inhibits oxidative stress and apoptosis via regulating SIRT1/NRF2 signaling, thereby reducing DOX-induced cardiotoxicity in H9c2 cells.
Collapse
Affiliation(s)
- 新华 李
- 西安医学院 药理学与毒理学教研室 (西安 710021)Department of Pharmacology and Toxicology, Xi'an Medical University, Xi'an 710021, China
| | - 爱丽 闫
- 西安医学院 药理学与毒理学教研室 (西安 710021)Department of Pharmacology and Toxicology, Xi'an Medical University, Xi'an 710021, China
| | - 晋瑞 常
- 西安医学院 药理学与毒理学教研室 (西安 710021)Department of Pharmacology and Toxicology, Xi'an Medical University, Xi'an 710021, China
| | - 汾 李
- 西安医学院 药理学与毒理学教研室 (西安 710021)Department of Pharmacology and Toxicology, Xi'an Medical University, Xi'an 710021, China
| | - 娟霞 朱
- 西安医学院 药理学与毒理学教研室 (西安 710021)Department of Pharmacology and Toxicology, Xi'an Medical University, Xi'an 710021, China
| |
Collapse
|
8
|
Bo L, Wang Y, Li Y, Wurpel JND, Huang Z, Chen ZS. The Battlefield of Chemotherapy in Pediatric Cancers. Cancers (Basel) 2023; 15:cancers15071963. [PMID: 37046624 PMCID: PMC10093214 DOI: 10.3390/cancers15071963] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/12/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023] Open
Abstract
The survival rate for pediatric cancers has remarkably improved in recent years. Conventional chemotherapy plays a crucial role in treating pediatric cancers, especially in low- and middle-income countries where access to advanced treatments may be limited. The Food and Drug Administration (FDA) approved chemotherapy drugs that can be used in children have expanded, but patients still face numerous side effects from the treatment. In addition, multidrug resistance (MDR) continues to pose a major challenge in improving the survival rates for a significant number of patients. This review focuses on the severe side effects of pediatric chemotherapy, including doxorubicin-induced cardiotoxicity (DIC) and vincristine-induced peripheral neuropathy (VIPN). We also delve into the mechanisms of MDR in chemotherapy to the improve survival and reduce the toxicity of treatment. Additionally, the review focuses on various drug transporters found in common types of pediatric tumors, which could offer different therapeutic options.
Collapse
Affiliation(s)
- Letao Bo
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY 11439, USA
| | - Youyou Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY 11439, USA
| | - Yidong Li
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY 11439, USA
| | - John N. D. Wurpel
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY 11439, USA
| | - Zoufang Huang
- Ganzhou Key Laboratory of Hematology, Department of Hematology, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
- Correspondence: (Z.H.); (Z.-S.C.); Tel.: +86-138-797-27439 (Z.H.); +1-718-990-1432 (Z.-S.C.); Fax: +1-718-990-1877 (Z.-S.C.)
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY 11439, USA
- Institute for Biotechnology, St. John’s University, Queens, NY 11439, USA
- Correspondence: (Z.H.); (Z.-S.C.); Tel.: +86-138-797-27439 (Z.H.); +1-718-990-1432 (Z.-S.C.); Fax: +1-718-990-1877 (Z.-S.C.)
| |
Collapse
|
9
|
van der Velden J, Asselbergs FW, Bakkers J, Batkai S, Bertrand L, Bezzina CR, Bot I, Brundel BJJM, Carrier L, Chamuleau S, Ciccarelli M, Dawson D, Davidson SM, Dendorfer A, Duncker DJ, Eschenhagen T, Fabritz L, Falcão-Pires I, Ferdinandy P, Giacca M, Girao H, Gollmann-Tepeköylü C, Gyongyosi M, Guzik TJ, Hamdani N, Heymans S, Hilfiker A, Hilfiker-Kleiner D, Hoekstra AG, Hulot JS, Kuster DWD, van Laake LW, Lecour S, Leiner T, Linke WA, Lumens J, Lutgens E, Madonna R, Maegdefessel L, Mayr M, van der Meer P, Passier R, Perbellini F, Perrino C, Pesce M, Priori S, Remme CA, Rosenhahn B, Schotten U, Schulz R, Sipido KR, Sluijter JPG, van Steenbeek F, Steffens S, Terracciano CM, Tocchetti CG, Vlasman P, Yeung KK, Zacchigna S, Zwaagman D, Thum T. Animal models and animal-free innovations for cardiovascular research: current status and routes to be explored. Consensus document of the ESC Working Group on Myocardial Function and the ESC Working Group on Cellular Biology of the Heart. Cardiovasc Res 2022; 118:3016-3051. [PMID: 34999816 PMCID: PMC9732557 DOI: 10.1093/cvr/cvab370] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 01/05/2022] [Indexed: 01/09/2023] Open
Abstract
Cardiovascular diseases represent a major cause of morbidity and mortality, necessitating research to improve diagnostics, and to discover and test novel preventive and curative therapies, all of which warrant experimental models that recapitulate human disease. The translation of basic science results to clinical practice is a challenging task, in particular for complex conditions such as cardiovascular diseases, which often result from multiple risk factors and comorbidities. This difficulty might lead some individuals to question the value of animal research, citing the translational 'valley of death', which largely reflects the fact that studies in rodents are difficult to translate to humans. This is also influenced by the fact that new, human-derived in vitro models can recapitulate aspects of disease processes. However, it would be a mistake to think that animal models do not represent a vital step in the translational pathway as they do provide important pathophysiological insights into disease mechanisms particularly on an organ and systemic level. While stem cell-derived human models have the potential to become key in testing toxicity and effectiveness of new drugs, we need to be realistic, and carefully validate all new human-like disease models. In this position paper, we highlight recent advances in trying to reduce the number of animals for cardiovascular research ranging from stem cell-derived models to in situ modelling of heart properties, bioinformatic models based on large datasets, and state-of-the-art animal models, which show clinically relevant characteristics observed in patients with a cardiovascular disease. We aim to provide a guide to help researchers in their experimental design to translate bench findings to clinical routine taking the replacement, reduction, and refinement (3R) as a guiding concept.
Collapse
Grants
- R01 HL150359 NHLBI NIH HHS
- RG/16/14/32397 British Heart Foundation
- FS/18/37/33642 British Heart Foundation
- PG/17/64/33205 British Heart Foundation
- PG/15/88/31780 British Heart Foundation
- FS/RTF/20/30009, NH/19/1/34595, PG/18/35/33786, CS/17/4/32960, PG/15/88/31780, and PG/17/64/33205 British Heart Foundation
- NC/T001488/1 National Centre for the Replacement, Refinement and Reduction of Animals in Research
- PG/18/44/33790 British Heart Foundation
- CH/16/3/32406 British Heart Foundation
- FS/RTF/20/30009 British Heart Foundation
- NWO-ZonMW
- ZonMW and Heart Foundation for the translational research program
- Dutch Cardiovascular Alliance (DCVA)
- Leducq Foundation
- Dutch Research Council
- Association of Collaborating Health Foundations (SGF)
- UCL Hospitals NIHR Biomedical Research Centre, and the DCVA
- Netherlands CardioVascular Research Initiative CVON
- Stichting Hartekind and the Dutch Research Counsel (NWO) (OCENW.GROOT.2019.029)
- National Fund for Scientific Research, Belgium and Action de Recherche Concertée de la Communauté Wallonie-Bruxelles, Belgium
- Netherlands CardioVascular Research Initiative CVON (PREDICT2 and CONCOR-genes projects), the Leducq Foundation
- ERA PerMed (PROCEED study)
- Netherlands Cardiovascular Research Initiative
- Dutch Heart Foundation
- German Centre of Cardiovascular Research (DZHH)
- Chest Heart and Stroke Scotland
- Tenovus Scotland
- Friends of Anchor and Grampian NHS-Endowments
- National Institute for Health Research University College London Hospitals Biomedical Research Centre
- German Centre for Cardiovascular Research
- European Research Council (ERC-AG IndivuHeart), the Deutsche Forschungsgemeinschaft
- European Union Horizon 2020 (REANIMA and TRAINHEART)
- German Ministry of Education and Research (BMBF)
- Centre for Cardiovascular Research (DZHK)
- European Union Horizon 2020
- DFG
- National Research, Development and Innovation Office of Hungary
- Research Excellence Program—TKP; National Heart Program
- Austrian Science Fund
- European Union Commission’s Seventh Framework programme
- CVON2016-Early HFPEF
- CVON She-PREDICTS
- CVON Arena-PRIME
- European Union’s Horizon 2020 research and innovation programme
- Deutsche Forschungsgemeinschaft
- Volkswagenstiftung
- French National Research Agency
- ERA-Net-CVD
- Fédération Française de Cardiologie, the Fondation pour la Recherche Médicale
- French PIA Project
- University Research Federation against heart failure
- Netherlands Heart Foundation
- Dekker Senior Clinical Scientist
- Health Holland TKI-LSH
- TUe/UMCU/UU Alliance Fund
- south African National Foundation
- Cancer Association of South Africa and Winetech
- Netherlands Heart Foundation/Applied & Engineering Sciences
- Dutch Technology Foundation
- Pie Medical Imaging
- Netherlands Organisation for Scientific Research
- Dr. Dekker Program
- Netherlands CardioVascular Research Initiative: the Dutch Heart Foundation
- Dutch Federation of University Medical Centres
- Netherlands Organization for Health Research and Development and the Royal Netherlands Academy of Sciences for the GENIUS-II project
- Netherlands Organization for Scientific Research (NWO) (VICI grant); the European Research Council
- Incyte s.r.l. and from Ministero dell’Istruzione, Università e Ricerca Scientifica
- German Center for Cardiovascular Research (Junior Research Group & Translational Research Project), the European Research Council (ERC Starting Grant NORVAS),
- Swedish Heart-Lung-Foundation
- Swedish Research Council
- National Institutes of Health
- Bavarian State Ministry of Health and Care through the research project DigiMed Bayern
- ERC
- ERA-CVD
- Dutch Heart Foundation, ZonMw
- the NWO Gravitation project
- Ministero dell'Istruzione, Università e Ricerca Scientifica
- Regione Lombardia
- Netherlands Organisation for Health Research and Development
- ITN Network Personalize AF: Personalized Therapies for Atrial Fibrillation: a translational network
- MAESTRIA: Machine Learning Artificial Intelligence Early Detection Stroke Atrial Fibrillation
- REPAIR: Restoring cardiac mechanical function by polymeric artificial muscular tissue
- Deutsche Forschungsgemeinschaft (DFG, German Research Foundation)
- European Union H2020 program to the project TECHNOBEAT
- EVICARE
- BRAV3
- ZonMw
- German Centre for Cardiovascular Research (DZHK)
- British Heart Foundation Centre for Cardiac Regeneration
- British Heart Foundation studentship
- NC3Rs
- Interreg ITA-AUS project InCARDIO
- Italian Association for Cancer Research
Collapse
Affiliation(s)
- Jolanda van der Velden
- Amsterdam UMC, Vrije Universiteit, Physiology, Amsterdam Cardiovascular Science, Amsterdam, The Netherlands
- Netherlands Heart Institute, Utrecht, The Netherlands
| | - Folkert W Asselbergs
- Division Heart & Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Faculty of Population Health Sciences, Institute of Cardiovascular Science and Institute of Health Informatics, University College London, London, UK
| | - Jeroen Bakkers
- Hubrecht Institute-KNAW and University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Sandor Batkai
- Hannover Medical School, Institute of Molecular and Translational Therapeutic Strategies, Hannover, Germany
| | - Luc Bertrand
- Hannover Medical School, Institute of Molecular and Translational Therapeutic Strategies, Hannover, Germany
| | - Connie R Bezzina
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Brussels, Belgium
| | - Ilze Bot
- Heart Center, Department of Experimental Cardiology, Amsterdam UMC, Location Academic Medical Center, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam, The Netherlands
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Bianca J J M Brundel
- Amsterdam UMC, Vrije Universiteit, Physiology, Amsterdam Cardiovascular Science, Amsterdam, The Netherlands
| | - Lucie Carrier
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Steven Chamuleau
- Amsterdam UMC, Heart Center, Cardiology, Amsterdam Cardiovascular Science, Amsterdam, The Netherlands
| | - Michele Ciccarelli
- Department of Medicine, Surgery and Odontology, University of Salerno, Fisciano (SA), Italy
| | - Dana Dawson
- Department of Cardiology, Aberdeen Cardiovascular and Diabetes Centre, Aberdeen Royal Infirmary and University of Aberdeen, Aberdeen, UK
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London WC1E 6HX, UK
| | - Andreas Dendorfer
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Dirk J Duncker
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Thomas Eschenhagen
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Larissa Fabritz
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
- University Center of Cardiovascular Sciences and Department of Cardiology, University Heart Center Hamburg, Germany and Institute of Cardiovascular Sciences, University of Birmingham, UK
| | - Ines Falcão-Pires
- UnIC - Cardiovascular Research and Development Centre, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Portugal
| | - Péter Ferdinandy
- Cardiometabolic Research Group and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Mauro Giacca
- Department of Medicine, Surgery and Health Sciences and Cardiovascular Department, Centre for Translational Cardiology, Azienda Sanitaria Universitaria Integrata Trieste, Trieste, Italy
- International Center for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
- King’s British Heart Foundation Centre, King’s College London, London, UK
| | - Henrique Girao
- Univ Coimbra, Center for Innovative Biomedicine and Biotechnology, Faculty of Medicine, Coimbra, Portugal
- Clinical Academic Centre of Coimbra, Coimbra, Portugal
| | | | - Mariann Gyongyosi
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Tomasz J Guzik
- Instutute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
- Jagiellonian University, Collegium Medicum, Kraków, Poland
| | - Nazha Hamdani
- Division Cardiology, Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
- Institute of Physiology, Ruhr University Bochum, Bochum, Germany
| | - Stephane Heymans
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht University, Maastricht, The Netherlands
- Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Andres Hilfiker
- Department for Cardiothoracic, Transplant, and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Denise Hilfiker-Kleiner
- Department for Cardiology and Angiology, Hannover Medical School, Hannover, Germany
- Department of Cardiovascular Complications in Pregnancy and in Oncologic Therapies, Comprehensive Cancer Centre, Philipps-Universität Marburg, Germany
| | - Alfons G Hoekstra
- Computational Science Lab, Informatics Institute, Faculty of Science, University of Amsterdam, Amsterdam, the Netherlands
| | - Jean-Sébastien Hulot
- Université de Paris, INSERM, PARCC, F-75015 Paris, France
- CIC1418 and DMU CARTE, AP-HP, Hôpital Européen Georges-Pompidou, F-75015 Paris, France
| | - Diederik W D Kuster
- Amsterdam UMC, Vrije Universiteit, Physiology, Amsterdam Cardiovascular Science, Amsterdam, The Netherlands
| | - Linda W van Laake
- Division Heart & Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Sandrine Lecour
- Department of Medicine, Hatter Institute for Cardiovascular Research in Africa and Cape Heart Institute, University of Cape Town, Cape Town, South Africa
| | - Tim Leiner
- Department of Radiology, Utrecht University Medical Center, Utrecht, the Netherlands
| | - Wolfgang A Linke
- Institute of Physiology II, University of Muenster, Robert-Koch-Str. 27B, 48149 Muenster, Germany
| | - Joost Lumens
- Department of Biomedical Engineering, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Esther Lutgens
- Experimental Vascular Biology Division, Department of Medical Biochemistry, University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München (LMU), Munich, Germany
- DZHK, Partner Site Munich Heart Alliance, Munich, Germany
| | - Rosalinda Madonna
- Department of Pathology, Cardiology Division, University of Pisa, 56124 Pisa, Italy
- Department of Internal Medicine, Cardiology Division, University of Texas Medical School in Houston, Houston, TX, USA
| | - Lars Maegdefessel
- DZHK, Partner Site Munich Heart Alliance, Munich, Germany
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Manuel Mayr
- King’s British Heart Foundation Centre, King’s College London, London, UK
| | - Peter van der Meer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Robert Passier
- Department of Applied Stem Cell Technologies, TechMed Centre, University of Twente, 7500AE Enschede, The Netherlands
- Department of Anatomy and Embryology, Leiden University Medical Centre, 2300 RC Leiden, The Netherlands
| | - Filippo Perbellini
- Hannover Medical School, Institute of Molecular and Translational Therapeutic Strategies, Hannover, Germany
| | - Cinzia Perrino
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Maurizio Pesce
- Unità di Ingegneria Tissutale Cardiovascolare, Centro cardiologico Monzino, IRCCS, Milan, Italy
| | - Silvia Priori
- Molecular Cardiology, Istituti Clinici Scientifici Maugeri, Pavia, Italy
- University of Pavia, Pavia, Italy
| | - Carol Ann Remme
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Brussels, Belgium
| | - Bodo Rosenhahn
- Institute for information Processing, Leibniz University of Hanover, 30167 Hannover, Germany
| | - Ulrich Schotten
- Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Rainer Schulz
- Institute of Physiology, Justus Liebig University Giessen, Giessen, Germany
| | - Karin R Sipido
- Department of Cardiovascular Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Joost P G Sluijter
- Experimental Cardiology Laboratory, Department of Cardiology, Regenerative Medicine Center Utrecht, Circulatory Health Laboratory, Utrecht University, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Frank van Steenbeek
- Division Heart & Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Sabine Steffens
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München (LMU), Munich, Germany
- DZHK, Partner Site Munich Heart Alliance, Munich, Germany
| | | | - Carlo Gabriele Tocchetti
- Cardio-Oncology Unit, Department of Translational Medical Sciences, Center for Basic and Clinical Immunology Research (CISI), Interdepartmental Center for Clinical and Translational Research (CIRCET), Interdepartmental Hypertension Research Center (CIRIAPA), Federico II University, Naples, Italy
| | - Patricia Vlasman
- Amsterdam UMC, Vrije Universiteit, Physiology, Amsterdam Cardiovascular Science, Amsterdam, The Netherlands
| | - Kak Khee Yeung
- Amsterdam UMC, Vrije Universiteit, Surgery, Amsterdam Cardiovascular Science, Amsterdam, The Netherlands
| | - Serena Zacchigna
- Department of Medicine, Surgery and Health Sciences and Cardiovascular Department, Centre for Translational Cardiology, Azienda Sanitaria Universitaria Integrata Trieste, Trieste, Italy
- International Center for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Dayenne Zwaagman
- Amsterdam UMC, Heart Center, Cardiology, Amsterdam Cardiovascular Science, Amsterdam, The Netherlands
| | - Thomas Thum
- Hannover Medical School, Institute of Molecular and Translational Therapeutic Strategies, Hannover, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| |
Collapse
|
10
|
Chen Y, Shi S, Dai Y. Research progress of therapeutic drugs for doxorubicin-induced cardiomyopathy. Biomed Pharmacother 2022; 156:113903. [PMID: 36279722 DOI: 10.1016/j.biopha.2022.113903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/16/2022] [Accepted: 10/19/2022] [Indexed: 12/06/2022] Open
|
11
|
Khairnar SI, Kulkarni YA, Singh K. Cardiotoxicity linked to anticancer agents and cardioprotective strategy. Arch Pharm Res 2022; 45:704-730. [DOI: 10.1007/s12272-022-01411-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 10/13/2022] [Indexed: 12/24/2022]
|
12
|
Zhang D, Lü J, Ren Z, Zhang X, Wu H, Sa R, Wang X, Wang Y, Lin Z, Zhang B. Potential cardiotoxicity induced by Euodiae Fructus: In vivo and in vitro experiments and untargeted metabolomics research. Front Pharmacol 2022; 13:1028046. [PMID: 36353487 PMCID: PMC9637925 DOI: 10.3389/fphar.2022.1028046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 10/05/2022] [Indexed: 09/16/2023] Open
Abstract
Background: Euodiae Fructus, a well-known herbal medicine, is widely used in Asia and has also gained in popularity in Western countries over the last decades. It has known side effects, which have been observed in clinical settings, but few studies have reported on its cardiotoxicity. Methods: In the present study, experiments using techniques of untargeted metabolomics clarify the hazardous effects of Euodiae Fructus on cardiac function and metabolism in rats in situations of overdosage and unsuitable syndrome differentiation. In vitro assays are conducted to observe the toxic effects of evodiamine and rutaecarpine, two main chemical constituents of Euodiae Fructus, in H9c2 and neonatal rat cardiomyocytes (NRCMs), with their signaling mechanisms analyzed accordingly. Results: The cardiac cytotoxicity of evodiamine and rutaecarpine in in vivo experiments is associated with remarkable alterations in lactate dehydrogenase, creatine kinase, and mitochondrial membrane potential; also with increased intensity of calcium fluorescence, decreased protein expression of the cGMP-PKG pathway in H9c2 cells, and frequency of spontaneous beat in NRCMs. Additionally, the results in rats with Yin deficiency receiving a high-dosage of Euodiae Fructus suggest obvious cardiac physiological dysfunction, abnormal electrocardiogram, pathological injuries, and decreased expression of PKG protein. At the level of endogenous metabolites, the cardiac side effects of overdose and irrational usage of Euodiae Fructus relate to 34 differential metabolites and 10 metabolic pathways involving among others, the purine metabolism, the glycerophospholipid metabolism, the glycerolipid metabolism, and the sphingolipid metabolism. Conclusion: These findings shed new light on the cardiotoxicity induced by Euodiae Fructus, which might be associated with overdose and unsuitable syndrome differentiation, that comes from modulating the cGMP-PKG pathway and disturbing the metabolic pathways of purine, lipid, and amino acid. Continuing research is needed to ensure pharmacovigilance for the safe administration of Chinese herbs in the future.
Collapse
Affiliation(s)
- Dan Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jintao Lü
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Zhixin Ren
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaomeng Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Centre for Pharmacovigilance and Rational Use of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Huanzhang Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Rina Sa
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Gansu Provincial Hospital, Lanzhou, China
| | - Xiaofang Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yu Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Centre for Pharmacovigilance and Rational Use of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Zhijian Lin
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Centre for Pharmacovigilance and Rational Use of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Bing Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Centre for Pharmacovigilance and Rational Use of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
13
|
Mineralocorticoid Receptor Pathway Is a Key Mediator of Carfilzomib-induced Nephrotoxicity: Preventive Role of Eplerenone. Hemasphere 2022; 6:e791. [PMID: 36285072 PMCID: PMC9584194 DOI: 10.1097/hs9.0000000000000791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 09/20/2022] [Indexed: 11/06/2022] Open
Abstract
Carfilzomib is an irreversible proteasome inhibitor indicated for relapsed/refractory multiple myeloma. Carfilzomib toxicity includes renal adverse effects (RAEs) of obscure pathobiology. Therefore, we investigated the mechanisms of nephrotoxicity developed by Carfilzomib. In a first experimental series, we used our previously established in vivo mouse models of Carfilzomib cardiotoxicity, that incorporated 2 and 4 doses of Carfilzomib, to identify whether Carfilzomib affects renal pathways. Hematology and biochemical analyses were performed, while kidneys underwent histological and molecular analyses. In a second and third experimental series, the 4 doses protocol was repeated for 24 hours urine collection and proteomic/metabolomic analyses. To test an experimental intervention, primary murine collecting duct tubular epithelial cells were treated with Carfilzomib and/or Eplerenone and Metformin. Finally, Eplerenone was orally co-administered with Carfilzomib daily (165 mg/kg) in the 4 doses protocol. We additionally used material from 7 patients to validate our findings and patients underwent biochemical analysis and assessment of renal mineralocorticoid receptor (MR) axis activation. In vivo screening showed that Carfilzomib-induced renal histological deficits and increased serum creatinine, urea, NGAL levels, and proteinuria only in the 4 doses protocol. Carfilzomib decreased diuresis, altered renal metabolism, and activated MR axis. This was consistent with the cytotoxicity found in primary murine collecting duct tubular epithelial cells, whereas Carfilzomib + Eplerenone co-administration abrogated Carfilzomib-related nephrotoxic effects in vitro and in vivo. Renal SGK-1, a marker of MR activation, increased in patients with Carfilzomib-related RAEs. Conclusively, Carfilzomib-induced renal MR/SGK-1 activation orchestrates RAEs and water retention both in vivo and in the clinical setting. MR blockade emerges as a potential therapeutic approach against Carfilzomib-related nephrotoxicity.
Collapse
|
14
|
Akhtar MS, Hassan MQ, Siddiqui A, Alavudeen SS, Afzal O, Altamimi ASA, Rahman SO, Khurana M, Ahsan MJ, Sharma AK, Tabassum F. Levosimendan: mechanistic insight and its diverse future aspects in cardiac care. Acta Cardiol 2022; 78:170-187. [PMID: 36222590 DOI: 10.1080/00015385.2022.2115761] [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: 11/01/2022]
Abstract
Inotropic agents are generally recommended to use in patients with acute decompensated heart failure (HF) with reduced ejection fraction (HFrEF) concurrent to end-organ dysfunction. However, due to certain pharmacological limitations like developing life threatening arrhythmia and tolerance, cannot be employed as much as needed. Meanwhile, Calcium ion (Ca2+) sensitisers exhibits their inotropic action by increasing the sensitivity of the cardiomyocyte to intracellular Ca2+ ion and have been reported as emerging therapeutic alternative in HF cases. Levosimendan (LEVO) is an inodilator and with its unique pharmacology justifying its use in a wide range of cardiac alterations in HF particularly in undergoing cardiac surgery. It is also reported to be better than classical inotropes in maintaining cardiac mechanical efficacy and reducing congestion in acute HF with hypotension. This review paper was designed to compile various evidence about basic pharmacology and potential clinical aspects of LEVO in cardiac surgery and other HF associated alterations. This will benefit directly to the researcher in initiating research and to fill the gaps in the area of thrust.
Collapse
Affiliation(s)
| | - Md Quamrul Hassan
- Department of Pharmacology, SNS College of Pharmacy, Motihari, India
| | - Aisha Siddiqui
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Selangor, Malaysia
| | | | - Obaid Afzal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Abdulmalik S A Altamimi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Syed Obaidur Rahman
- Department of Pharmacology, School of Pharmaceutical Education and Research, New Delhi, India
| | - Mallika Khurana
- Department of Pharmacology, School of Pharmaceutical Education and Research, New Delhi, India
| | - Mohamed Jawed Ahsan
- Department of Pharmaceutical Chemistry, Maharishi Arvind College of Pharmacy, Jaipur, India
| | - Arun Kumar Sharma
- Department of Pharmacology, Amity Institute of Pharmacy, Noida, India
| | - Fauzia Tabassum
- Department of Pharmacology, College of Dentistry and Pharmacy, Buraydah, Saudi Arabia
| |
Collapse
|
15
|
Molecular mechanisms associated with the chemoprotective role of protocatechuic acid and its potential benefits in the amelioration of doxorubicin-induced cardiotoxicity: A review. Toxicol Rep 2022; 9:1713-1724. [PMID: 36561952 PMCID: PMC9764176 DOI: 10.1016/j.toxrep.2022.09.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 09/01/2022] [Accepted: 09/03/2022] [Indexed: 12/25/2022] Open
Abstract
Since its discovery in the 1960 s, doxorubicin (DOX) has constantly elicited the broadest spectrum of cancerocidal activity against human cancers. However, cardiotoxicity caused by DOX directly as well as its metabolites is a great source of concern over the continuous use of DOX in chemotherapy. While the exact mechanism of DOX-induced cardiotoxicity is yet to be completely understood, recent studies indicate oxidative stress, inflammation, and several forms of cell death as key pathogenic mechanisms that underpin the etiology of doxorubicin-induced cardiotoxicity (DIC). Notably, these key mechanistic events are believed to be negatively regulated by 3,4-dihydroxybenzoic acid or protocatechuic acid (PCA)-a plant-based phytochemical with proven anti-oxidant, anti-inflammatory, and anti-apoptotic properties. Here, we review the experimental findings detailing the potential ameliorative effects of PCA under exposure to DOX. We also discuss molecular insights into the pathophysiology of DIC, highlighting the potential intervention points where the use of PCA as a veritable chemoprotective agent may ameliorate DOX-induced cardiotoxicities as well as toxicities due to other anticancer drugs like cisplatin. While we acknowledge that controlled oral administration of PCA during chemotherapy may be insufficient to eliminate all toxicities due to DOX treatment, we propose that the ability of PCA to block oxidative stress, attenuate inflammation, and abrogate several forms of cardiomyocyte cell death underlines its great promise in the amelioration of DIC.
Collapse
|
16
|
Wang Z, Bian W, Yan Y, Zhang DM. Functional Regulation of KATP Channels and Mutant Insight Into Clinical Therapeutic Strategies in Cardiovascular Diseases. Front Pharmacol 2022; 13:868401. [PMID: 35837280 PMCID: PMC9274113 DOI: 10.3389/fphar.2022.868401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 06/03/2022] [Indexed: 11/13/2022] Open
Abstract
ATP-sensitive potassium channels (KATP channels) play pivotal roles in excitable cells and link cellular metabolism with membrane excitability. The action potential converts electricity into dynamics by ion channel-mediated ion exchange to generate systole, involved in every heartbeat. Activation of the KATP channel repolarizes the membrane potential and decreases early afterdepolarization (EAD)-mediated arrhythmias. KATP channels in cardiomyocytes have less function under physiological conditions but they open during severe and prolonged anoxia due to a reduced ATP/ADP ratio, lessening cellular excitability and thus preventing action potential generation and cell contraction. Small active molecules activate and enhance the opening of the KATP channel, which induces the repolarization of the membrane and decreases the occurrence of malignant arrhythmia. Accumulated evidence indicates that mutation of KATP channels deteriorates the regulatory roles in mutation-related diseases. However, patients with mutations in KATP channels still have no efficient treatment. Hence, in this study, we describe the role of KATP channels and subunits in angiocardiopathy, summarize the mutations of the KATP channels and the functional regulation of small active molecules in KATP channels, elucidate the potential mechanisms of mutant KATP channels and provide insight into clinical therapeutic strategies.
Collapse
Affiliation(s)
- Zhicheng Wang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Weikang Bian
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yufeng Yan
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Dai-Min Zhang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- Department of Cardiology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
- *Correspondence: Dai-Min Zhang,
| |
Collapse
|
17
|
Kong CY, Guo Z, Song P, Zhang X, Yuan YP, Teng T, Yan L, Tang QZ. Underlying the Mechanisms of Doxorubicin-Induced Acute Cardiotoxicity: Oxidative Stress and Cell Death. Int J Biol Sci 2022; 18:760-770. [PMID: 35002523 PMCID: PMC8741835 DOI: 10.7150/ijbs.65258] [Citation(s) in RCA: 139] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 11/17/2021] [Indexed: 02/06/2023] Open
Abstract
Cancer is a destructive disease that causes high levels of morbidity and mortality. Doxorubicin (DOX) is a highly efficient antineoplastic chemotherapeutic drug, but its use places survivors at risk for cardiotoxicity. Many studies have demonstrated that multiple factors are involved in DOX-induced acute cardiotoxicity. Among them, oxidative stress and cell death predominate. In this review, we provide a comprehensive overview of the mechanisms underlying the source and effect of free radicals and dependent cell death pathways induced by DOX. Hence, we attempt to explain the cellular mechanisms of oxidative stress and cell death that elicit acute cardiotoxicity and provide new insights for researchers to discover potential therapeutic strategies to prevent or reverse doxorubicin-induced cardiotoxicity.
Collapse
Affiliation(s)
- Chun-Yan Kong
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, RP China
| | - Zhen Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, RP China
| | - Peng Song
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, RP China
| | - Xin Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, RP China
| | - Yu-Pei Yuan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, RP China
| | - Teng Teng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, RP China
| | - Ling Yan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, RP China
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, RP China
| |
Collapse
|
18
|
Elucidating Carfilzomib's Induced Cardiotoxicity in an In Vivo Model of Aging: Prophylactic Potential of Metformin. Int J Mol Sci 2021; 22:ijms222010956. [PMID: 34681615 PMCID: PMC8537073 DOI: 10.3390/ijms222010956] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/04/2021] [Accepted: 10/04/2021] [Indexed: 02/07/2023] Open
Abstract
Background: Carfilzomib is a first-line proteasome inhibitor indicated for relapsed/refractory multiple myeloma (MM), with its clinical use being hampered by cardiotoxic phenomena. We have previously established a translational model of carfilzomib cardiotoxicity in young adult mice, in which metformin emerged as a prophylactic therapy. Considering that MM is an elderly disease and that age is an independent risk factor for cardiotoxicity, herein, we sought to validate carfilzomib’s cardiotoxicity in an in vivo model of aging. Methods: Aged mice underwent the translational two- and four-dose protocols without and with metformin. Mice underwent echocardiography and were subsequently sacrificed for molecular analyses in the blood and cardiac tissue. Results: Carfilzomib decreased proteasomal activity both in PBMCs and myocardium in both protocols. Carfilzomib induced mild cardiotoxicity after two doses and more pronounced cardiomyopathy in the four-dose protocol, while metformin maintained cardiac function. Carfilzomib led to an increased Bip expression and decreased AMPKα phosphorylation, while metformin coadministration partially decreased Bip expression and induced AMPKα phosphorylation, leading to enhanced myocardial LC3B-dependent autophagy. Conclusion: Carfilzomib induced cardiotoxicity in aged mice, an effect significantly reversed by metformin. The latter possesses translational importance as it further supports the clinical use of metformin as a potent prophylactic therapy.
Collapse
|
19
|
Armandeh M, Bameri B, Baeeri M, Haghi-Aminjan H, Rahimifard M, Hassani S, Hooshangi Shayesteh MR, Khalid M, Samadi M, Hosseini R, Masoudi Fard M, Abdollahi M. The role of levosimendan in phosphine-induced cardiotoxicity: evaluation of electrocardiographic, echocardiographic, and biochemical parameters. Toxicol Mech Methods 2021; 31:631-643. [PMID: 34219611 DOI: 10.1080/15376516.2021.1950248] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Aluminum phosphide (AlP) causes serious poisoning in which severe cardiac suppression is the significant lethal consequence. According to evidence, levosimendan can exert outstanding cardiac support and protection in different pathological conditions. This study aimed to investigate the mechanisms by which levosimendan may alleviate cardiovascular toxicity due to AlP intoxication in the rat model. The groups included control group (normal saline only), sole levosimendan groups (12, 24, 48 μg/kg), AlP group (10 mg/kg), and AlP + levosimendan groups receiving 12, 24, 48 μg/kg levosimendan intraperitoneally 30 min after AlP administration. Electrocardiographic (ECG) parameters (QRS and PR duration and ST height), heart rate, and blood pressure were monitored for 180 minutes. Also, after 24 h of poisoning, echocardiography was applied to assess left ventricle function. Evaluation of the biochemical parameters in heart tissue, including mitochondrial complexes I, II, IV activity, ADP/ATP ratio, the rate of apoptosis, malondialdehyde (MDA), lactate, and troponin I levels, were done after 12 and 24 h. AlP-induced ECG abnormalities (PR duration and ST height), reduction in heart rate, blood pressure, cardiac output, ejection fraction, and stroke volume were improved by levosimendan administration. Besides, levosimendan significantly improved complex IV activity, the ADP/ATP ratio, apoptosis, MDA, lactate, and troponin I level following AlP-poisoning. Our results suggest that levosimendan might alleviate AlP-induced cardiotoxicity by modulating mitochondrial activity and improving cardiac function. However, the potential clinical use of levosimendan in this toxicity needs more investigations.
Collapse
Affiliation(s)
- Maryam Armandeh
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.,Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (P SRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Behnaz Bameri
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.,Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (P SRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Baeeri
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.,Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (P SRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Hamed Haghi-Aminjan
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mahban Rahimifard
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.,Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (P SRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Shokoufeh Hassani
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.,Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (P SRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Hooshangi Shayesteh
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.,Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (P SRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Madiha Khalid
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.,Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (P SRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Mahedeh Samadi
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rohollah Hosseini
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Majid Masoudi Fard
- Department of Surgery & Radiology, Faculty of Veterinary Medicine, Tehran University, Tehran, Iran
| | - Mohammad Abdollahi
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.,Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (P SRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
20
|
Efentakis P, Molitor M, Kossmann S, Bochenek ML, Wild J, Lagrange J, Finger S, Jung R, Karbach S, Schäfer K, Schulz A, Wild P, Münzel T, Wenzel P. Tubulin-folding cofactor E deficiency promotes vascular dysfunction by increased endoplasmic reticulum stress. Eur Heart J 2021; 43:488-500. [PMID: 34132336 PMCID: PMC8830526 DOI: 10.1093/eurheartj/ehab222] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 10/29/2020] [Accepted: 03/26/2021] [Indexed: 12/14/2022] Open
Abstract
AIMS Assessment of endothelial function in humans by measuring flow-mediated dilation (FMD) risk-stratifies individuals with established cardiovascular disease, whereas its predictive value is limited in primary prevention. We therefore aimed to establish and evaluate novel markers of FMD at the population level. METHODS AND RESULTS In order to identify novel targets that were negatively correlated with FMD and investigate their contribution to vascular function, we performed a genome-wide association study (GWAS) of 4175 participants of the population based Gutenberg Health Study. Subsequently, conditional knockout mouse models deleting the gene of interest were generated and characterized. GWAS analysis revealed that single-nucleotide polymorphisms (SNPs) in the tubulin-folding cofactor E (TBCE) gene were negatively correlated with endothelial function and TBCE expression. Vascular smooth muscle cell (VSMC)-targeted TBCE deficiency was associated with endothelial dysfunction, aortic wall hypertrophy, and endoplasmic reticulum (ER) stress-mediated VSMC hyperproliferation in mice, paralleled by calnexin up-regulation and exacerbated by the blood pressure hormone angiotensin II. Treating SMMHC-ERT2-Cre+/-TBCEfl/fl mice with the ER stress modulator tauroursodeoxycholic acid amplified Raptor/Beclin-1-dependent autophagy and reversed vascular dysfunction. CONCLUSION TBCE and tubulin homeostasis seem to be novel predictors of vascular function and offer a new drug target to ameliorate ER stress-dependent vascular dysfunction.
Collapse
Affiliation(s)
- Panagiotis Efentakis
- Department of Cardiology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany.,Center for Thrombosis and Hemostasis, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Michael Molitor
- Department of Cardiology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany.,Center for Thrombosis and Hemostasis, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany.,German Center for Cardiovascular Research (DZHK)-Partner site Rhine-Main, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Sabine Kossmann
- Department of Cardiology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany.,Center for Thrombosis and Hemostasis, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Magdalena L Bochenek
- Department of Cardiology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany.,Center for Thrombosis and Hemostasis, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany.,German Center for Cardiovascular Research (DZHK)-Partner site Rhine-Main, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Johannes Wild
- Department of Cardiology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany.,Center for Thrombosis and Hemostasis, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Jeremy Lagrange
- Department of Cardiology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany.,Center for Thrombosis and Hemostasis, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Stefanie Finger
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Rebecca Jung
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Susanne Karbach
- Department of Cardiology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany.,Center for Thrombosis and Hemostasis, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany.,German Center for Cardiovascular Research (DZHK)-Partner site Rhine-Main, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Katrin Schäfer
- Department of Cardiology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany.,German Center for Cardiovascular Research (DZHK)-Partner site Rhine-Main, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Andreas Schulz
- Department of Cardiology-Preventive Cardiology and Medical Prevention, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Philipp Wild
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany.,German Center for Cardiovascular Research (DZHK)-Partner site Rhine-Main, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany.,Department of Cardiology-Preventive Cardiology and Medical Prevention, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Thomas Münzel
- Department of Cardiology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany.,Center for Thrombosis and Hemostasis, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany.,German Center for Cardiovascular Research (DZHK)-Partner site Rhine-Main, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Philip Wenzel
- Department of Cardiology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany.,Center for Thrombosis and Hemostasis, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany.,German Center for Cardiovascular Research (DZHK)-Partner site Rhine-Main, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| |
Collapse
|
21
|
Attanasio U, Pirozzi F, Poto R, Cuomo A, Carannante A, Russo M, Ghigo A, Hirsch E, Tocchetti CG, Varricchi G, Mercurio V. Oxidative stress in anticancer therapies-related cardiac dysfunction. Free Radic Biol Med 2021; 169:410-415. [PMID: 33930514 DOI: 10.1016/j.freeradbiomed.2021.04.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 03/30/2021] [Accepted: 04/08/2021] [Indexed: 02/06/2023]
Abstract
Redox abnormalities are at the crossroad of cardiovascular diseases, cancer and cardiotoxicity from anticancer treatments. Indeed, disturbances of the redox equilibrium are common drivers of these conditions. Not only is an increase in oxidative stress a fundamental mechanism of action of anthracyclines (which have historically been the most studied anticancer treatments) but also this is at the basis of the toxic cardiovascular effects of antineoplastic targeted drugs and radiotherapy. Here we examine the oxidative mechanisms involved in the different cardiotoxicities induced by the main redox-based antineoplastic treatments, and discuss novel approaches for the treatment of such toxicities.
Collapse
Affiliation(s)
- Umberto Attanasio
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Flora Pirozzi
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Remo Poto
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Alessandra Cuomo
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Antonio Carannante
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Michele Russo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Carlo Gabriele Tocchetti
- Department of Translational Medical Sciences, Federico II University, Naples, Italy; Interdepartmental Center of Clinical and Translational Research (CIRCET), Federico II University, Naples, Italy; Interdepartmental Hypertension Research Center (CIRIAPA), Federico II University, Naples, Italy; Center for Basic and Clinical Immunology Research (CISI), Federico II University, Naples, Italy.
| | - Gilda Varricchi
- Department of Translational Medical Sciences, Federico II University, Naples, Italy; Center for Basic and Clinical Immunology Research (CISI), Federico II University, Naples, Italy; WAO Center of Excellence, Naples, Italy; Institute of Experimental Endocrinology and Oncology "G. Salvatore" (IEOS), National Research Council (CNR), Naples, Italy
| | - Valentina Mercurio
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| |
Collapse
|
22
|
Zhang Y, Ni L, Lin B, Hu L, Lin Z, Yang J, Wang J, Ma H, Liu Y, Yang J, Lin J, Xu L, Wu L, Shi D. SNX17 protects the heart from doxorubicin-induced cardiotoxicity by modulating LMOD2 degradation. Pharmacol Res 2021; 169:105642. [PMID: 33933636 DOI: 10.1016/j.phrs.2021.105642] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 04/17/2021] [Accepted: 04/22/2021] [Indexed: 12/25/2022]
Abstract
Anthracyclines including doxorubicin (DOX) are still the most widely used and efficacious antitumor drugs, although their cardiotoxicity is a significant cause of heart failure. Despite considerable efforts being made to minimize anthracycline-induced cardiac adverse effects, little progress has been achieved. In this study, we aimed to explore the role and underlying mechanism of SNX17 in DOX-induced cardiotoxicity. We found that SNX17 was downregulated in cardiomyocytes treated with DOX both in vitro and in vivo. DOX treatment combined with SNX17 interference worsened the damage to neonatal rat ventricular myocytes (NRVMs). Furthermore, the rats with SNX17 deficiency manifested increased susceptibility to DOX-induced cardiotoxicity (myocardial damage and fibrosis, impaired contractility and cardiac death). Mechanistic investigation revealed that SNX17 interacted with leiomodin-2 (LMOD2), a key regulator of the thin filament length in muscles, via its C-TERM domain and SNX17 deficiency exacerbated DOX-induced cardiac systolic dysfunction by promoting aberrant LMOD2 degradation through lysosomal pathway. In conclusion, these findings highlight that SNX17 plays a protective role in DOX-induced cardiotoxicity, which provides an attractive target for the prevention and treatment of anthracycline induced cardiotoxicity.
Collapse
Affiliation(s)
- Yanping Zhang
- Department of Vascular and Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Le Ni
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Bowen Lin
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Lingjie Hu
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Zheyi Lin
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Jian Yang
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Jinyu Wang
- Department of Physiology, Shanxi Medical University, Taiyuan 030001, China
| | - Honghui Ma
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Yi Liu
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Jian Yang
- Jinzhou Medical University, Liaoning 121000, China
| | - Jianghua Lin
- Jinzhou Medical University, Liaoning 121000, China
| | - Liang Xu
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Liqun Wu
- Department of Vascular and Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Dan Shi
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China.
| |
Collapse
|
23
|
Roth S, Torregroza C, Feige K, Preckel B, Hollmann MW, Weber NC, Huhn R. Pharmacological Conditioning of the Heart: An Update on Experimental Developments and Clinical Implications. Int J Mol Sci 2021; 22:ijms22052519. [PMID: 33802308 PMCID: PMC7959135 DOI: 10.3390/ijms22052519] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/26/2021] [Accepted: 02/26/2021] [Indexed: 12/13/2022] Open
Abstract
The aim of pharmacological conditioning is to protect the heart against myocardial ischemia-reperfusion (I/R) injury and its consequences. There is extensive literature that reports a multitude of different cardioprotective signaling molecules and mechanisms in diverse experimental protocols. Several pharmacological agents have been evaluated in terms of myocardial I/R injury. While results from experimental studies are immensely encouraging, translation into the clinical setting remains unsatisfactory. This narrative review wants to focus on two aspects: (1) give a comprehensive update on new developments of pharmacological conditioning in the experimental setting concentrating on recent literature of the last two years and (2) briefly summarize clinical evidence of these cardioprotective substances in the perioperative setting highlighting their clinical implications. By directly opposing each pharmacological agent regarding its recent experimental knowledge and most important available clinical data, a clear overview is given demonstrating the remaining gap between basic research and clinical practice. Finally, future perspectives are given on how we might overcome the limited translatability in the field of pharmacological conditioning.
Collapse
Affiliation(s)
- Sebastian Roth
- Department of Anesthesiology, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University Duesseldorf, Moorenstr. 5, 40225 Duesseldorf, Germany; (S.R.); (K.F.); (R.H.)
| | - Carolin Torregroza
- Department of Anesthesiology, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University Duesseldorf, Moorenstr. 5, 40225 Duesseldorf, Germany; (S.R.); (K.F.); (R.H.)
- Department of Anesthesiology, Amsterdam University Medical Center (AUMC), Location AMC, Meibergdreef 9, 1100 DD Amsterdam, The Netherlands; (B.P.); (M.W.H.); (N.C.W.)
- Correspondence:
| | - Katharina Feige
- Department of Anesthesiology, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University Duesseldorf, Moorenstr. 5, 40225 Duesseldorf, Germany; (S.R.); (K.F.); (R.H.)
| | - Benedikt Preckel
- Department of Anesthesiology, Amsterdam University Medical Center (AUMC), Location AMC, Meibergdreef 9, 1100 DD Amsterdam, The Netherlands; (B.P.); (M.W.H.); (N.C.W.)
| | - Markus W. Hollmann
- Department of Anesthesiology, Amsterdam University Medical Center (AUMC), Location AMC, Meibergdreef 9, 1100 DD Amsterdam, The Netherlands; (B.P.); (M.W.H.); (N.C.W.)
| | - Nina C. Weber
- Department of Anesthesiology, Amsterdam University Medical Center (AUMC), Location AMC, Meibergdreef 9, 1100 DD Amsterdam, The Netherlands; (B.P.); (M.W.H.); (N.C.W.)
| | - Ragnar Huhn
- Department of Anesthesiology, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University Duesseldorf, Moorenstr. 5, 40225 Duesseldorf, Germany; (S.R.); (K.F.); (R.H.)
| |
Collapse
|
24
|
Nikolaou PE, Efentakis P, Abu Qourah F, Femminò S, Makridakis M, Kanaki Z, Varela A, Tsoumani M, Davos CH, Dimitriou CA, Tasouli A, Dimitriadis G, Kostomitsopoulos N, Zuurbier CJ, Vlahou A, Klinakis A, Brizzi MF, Iliodromitis EK, Andreadou I. Chronic Empagliflozin Treatment Reduces Myocardial Infarct Size in Nondiabetic Mice Through STAT-3-Mediated Protection on Microvascular Endothelial Cells and Reduction of Oxidative Stress. Antioxid Redox Signal 2021; 34:551-571. [PMID: 32295413 DOI: 10.1089/ars.2019.7923] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Aims: Empagliflozin (EMPA) demonstrates cardioprotective effects on diabetic myocardium but its infarct-sparing effects in normoglycemia remain unspecified. We investigated the acute and chronic effect of EMPA on infarct size after ischemia-reperfusion (I/R) injury and the mechanisms of cardioprotection in nondiabetic mice. Results: Chronic oral administration of EMPA (6 weeks) reduced myocardial infarct size after 30 min/2 h I/R (26.5% ± 3.9% vs 45.8% ± 3.3% in the control group, p < 0.01). Body weight, blood pressure, glucose levels, and cardiac function remained unchanged between groups. Acute administration of EMPA 24 or 4 h before I/R did not affect infarct size. Chronic EMPA treatment led to a significant reduction of oxidative stress biomarkers. STAT-3 (signal transducer and activator of transcription 3) was activated by Y(705) phosphorylation at the 10th minute of R, but it remained unchanged at 2 h of R and in the acute administration protocols. Proteomic analysis was employed to investigate signaling intermediates and revealed that chronic EMPA treatment regulates several pathways at reperfusion, including oxidative stress and integrin-related proteins that were further evaluated. Superoxide dismutase and vascular endothelial growth factor were increased throughout reperfusion. EMPA pretreatment (24 h) increased the viability of human microvascular endothelial cells in normoxia and on 3 h hypoxia/1 h reoxygenation and reduced reactive oxygen species production. In EMPA-treated murine hearts, CD31-/VEGFR2-positive endothelial cells and the pSTAT-3(Y705) signal derived from endothelial cells were boosted at early reperfusion. Innovation: Chronic EMPA administration reduces infarct size in healthy mice via the STAT-3 pathway and increases the survival of endothelial cells. Conclusion: Chronic but not acute administration of EMPA reduces infarct size through STAT-3 activation independently of diabetes mellitus.
Collapse
Affiliation(s)
| | - Panagiotis Efentakis
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Fairouz Abu Qourah
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Saveria Femminò
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Manousos Makridakis
- Biotechnology Laboratory, Centre of Basic Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece
| | - Zoi Kanaki
- Biomedical Research Foundation Academy of Athens, Athens, Greece
| | - Aimilia Varela
- Cardiovascular Research Laboratory, Biomedical Research Foundation Academy of Athens, Athens, Greece
| | - Maria Tsoumani
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Constantinos H Davos
- Cardiovascular Research Laboratory, Biomedical Research Foundation Academy of Athens, Athens, Greece
| | - Constantinos A Dimitriou
- Cardiovascular Research Laboratory, Biomedical Research Foundation Academy of Athens, Athens, Greece
| | | | - George Dimitriadis
- 2nd Department of Internal Medicine, Research Institute and Diabetes Center, National and Kapodistrian University of Athens, "Attikon" University Hospital, Athens, Greece
| | - Nikolaos Kostomitsopoulos
- Academy of Athens Biomedical Research Foundation, Centre of Clinical Experimental Surgery and Translational Research, Athens, Greece
| | - Coert J Zuurbier
- Amsterdam UMC, University of Amsterdam, Laboratory of Experimental Intensive Care and Anesthesiology, Department of Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam Infection & Immunity, Amsterdam, The Netherlands
| | - Antonia Vlahou
- Biotechnology Laboratory, Centre of Basic Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece
| | | | - Maria F Brizzi
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Efstathios K Iliodromitis
- 2nd University Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
25
|
Liu Y, Chen J, Fontes SK, Bautista EN, Cheng Z. Physiological And Pathological Roles Of Protein Kinase A In The Heart. Cardiovasc Res 2021; 118:386-398. [PMID: 33483740 DOI: 10.1093/cvr/cvab008] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/30/2020] [Accepted: 01/08/2021] [Indexed: 12/21/2022] Open
Abstract
Protein kinase A (PKA) is a central regulator of cardiac performance and morphology. Myocardial PKA activation is induced by a variety of hormones, neurotransmitters and stress signals, most notably catecholamines secreted by the sympathetic nervous system. Catecholamines bind β-adrenergic receptors to stimulate cAMP-dependent PKA activation in cardiomyocytes. Elevated PKA activity enhances Ca2+ cycling and increases cardiac muscle contractility. Dynamic control of PKA is essential for cardiac homeostasis, as dysregulation of PKA signaling is associated with a broad range of heart diseases. Specifically, abnormal PKA activation or inactivation contributes to the pathogenesis of myocardial ischemia, hypertrophy, heart failure, as well as diabetic, takotsubo, or anthracycline cardiomyopathies. PKA may also determine sex-dependent differences in contractile function and heart disease predisposition. Here, we describe the recent advances regarding the roles of PKA in cardiac physiology and pathology, highlighting previous study limitations and future research directions. Moreover, we discuss the therapeutic strategies and molecular mechanisms associated with cardiac PKA biology. In summary, PKA could serve as a promising drug target for cardioprotection. Depending on disease types and mechanisms, therapeutic intervention may require either inhibition or activation of PKA. Therefore, specific PKA inhibitors or activators may represent valuable drug candidates for the treatment of heart diseases.
Collapse
Affiliation(s)
- Yuening Liu
- Department of Pharmaceutical Sciences, Washington State University, PBS 423, 412 E. Spokane Falls Blvd, ., Spokane, WA, 99202-2131, USA
| | - Jingrui Chen
- Department of Pharmaceutical Sciences, Washington State University, PBS 423, 412 E. Spokane Falls Blvd, ., Spokane, WA, 99202-2131, USA
| | - Shayne K Fontes
- Department of Pharmaceutical Sciences, Washington State University, PBS 423, 412 E. Spokane Falls Blvd, ., Spokane, WA, 99202-2131, USA
| | - Erika N Bautista
- Department of Pharmaceutical Sciences, Washington State University, PBS 423, 412 E. Spokane Falls Blvd, ., Spokane, WA, 99202-2131, USA
| | - Zhaokang Cheng
- Department of Pharmaceutical Sciences, Washington State University, PBS 423, 412 E. Spokane Falls Blvd, ., Spokane, WA, 99202-2131, USA
| |
Collapse
|
26
|
Saiyang X, Qingqing W, man X, Chen L, Min Z, Yun X, Wenke S, Haiming W, Xiaofeng Z, Si C, Haipeng G, Wei D, Qizhu T. Activation of Toll-like receptor 7 provides cardioprotection in septic cardiomyopathy-induced systolic dysfunction. Clin Transl Med 2021; 11:e266. [PMID: 33463061 PMCID: PMC7775988 DOI: 10.1002/ctm2.266] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 12/07/2020] [Accepted: 12/12/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND As a pattern recognition receptor, Toll-like receptor 7 (TLR7) widely presented in the endosomal membrane of various cells. However, the precise role and mechanism of TLR7 in septic cardiomyopathy remain unknown. This study aims to determine the role of TLR7 in cardiac dysfunction during sepsis and explore the mechanism of TLR7 in septic cardiomyopathy. METHODS We generated a mouse model of septic cardiomyopathy by challenging with lipopolysaccharide (LPS). TLR7-knockout (TLR7-/- ), wild-type (WT) mice, cardiac-specific TLR7-transgenic (cTG-TLR7) overexpression, and littermates WT (LWT) mice were subjected to septic model. Additionally, to verify the role and mechanism of TLR7 in vitro, we transfected neonatal rat ventricular myocytes (NRVMs) with Ad-TLR7 and TLR7 siRNA before LPS administration. The effects of TLR7 were assessed by Ca2+ imaging, western blotting, immunostaining, and quantitative real-time polymerase chain reaction (qPCR). RESULTS We found that TLR7 knockout markedly exacerbated sepsis-induced systolic dysfunction. Moreover, cardiomyocytes isolated from TLR7-/- mice displayed weaker Ca2+ handling than that in WT mice in response to LPS. Conversely, TLR7 overexpression alleviated LPS-induced systolic dysfunction, and loxoribine (TLR7-specific agonist) improved LPS-induced cardiac dysfunction. Mechanistically, these optimized effects were associated with enhanced the adenosine (cAMP)-protein kinase A (PKA) pathway, which upregulated phosphorylate-phospholamban (p-PLN) (Ser16) and promoted sarco/endoplasmic reticulum Ca2+ ATPase (Serca) and Ryanodine Receptor 2 (RyR2) expression in the sarcoplasmic reticulum (SR), and ultimately restored Ca2+ handling in response to sepsis. While improved Ca2+ handling was abrogated after H89 (a specific PKA inhibitor) pretreatment in cardiomyocytes isolated from cTG-TLR7 mice. Consistently, TLR7 overexpression improved LPS-induced Ca2+ -handling decrement in NRVMs. Nevertheless, TLR7 knockdown showed a deteriorative phenotype. CONCLUSIONS Our data demonstrated that activation of TLR7 protected against sepsis-induced cardiac dysfunction through promoting cAMP-PKA-PLN pathway, and we revealed that TLR7 might be a novel therapeutic target to block the septic cardiomyopathy and support systolic function during sepsis.
Collapse
Affiliation(s)
- Xie Saiyang
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanPeople's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic DiseasesWuhanPeople's Republic of China
| | - Wu Qingqing
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanPeople's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic DiseasesWuhanPeople's Republic of China
| | - Xu man
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanPeople's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic DiseasesWuhanPeople's Republic of China
| | - Liu Chen
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanPeople's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic DiseasesWuhanPeople's Republic of China
| | - Zhang Min
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanPeople's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic DiseasesWuhanPeople's Republic of China
| | - Xing Yun
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanPeople's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic DiseasesWuhanPeople's Republic of China
| | - Shi Wenke
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanPeople's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic DiseasesWuhanPeople's Republic of China
| | - Wu Haiming
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanPeople's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic DiseasesWuhanPeople's Republic of China
| | - Zeng Xiaofeng
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanPeople's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic DiseasesWuhanPeople's Republic of China
| | - Chen Si
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanPeople's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic DiseasesWuhanPeople's Republic of China
| | - Guo Haipeng
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of HealthQilu Hospital of Shandong UniversityJinanChina
- Department of Critical Care MedicineQilu Hospital of Shandong UniversityJinanPeople's Republic of China
| | - Deng Wei
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanPeople's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic DiseasesWuhanPeople's Republic of China
- Department of CardiologyThe Fifth Affiliated Hospital of Xinjiang Medical UniversityÜrümqiChina
| | - Tang Qizhu
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanPeople's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic DiseasesWuhanPeople's Republic of China
| |
Collapse
|
27
|
Tanaka Y, Nagoshi T, Yoshii A, Oi Y, Takahashi H, Kimura H, Ito K, Kashiwagi Y, Tanaka TD, Yoshimura M. Xanthine oxidase inhibition attenuates doxorubicin-induced cardiotoxicity in mice. Free Radic Biol Med 2021; 162:298-308. [PMID: 33470212 DOI: 10.1016/j.freeradbiomed.2020.10.303] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 10/15/2020] [Accepted: 10/20/2020] [Indexed: 12/20/2022]
Abstract
Accumulating evidence suggests that high serum uric acid (UA) is associated with left ventricular (LV) dysfunction. Although xanthine oxidase (XO) activation is a critical regulatory mechanism of the terminal step in ATP and purine degradation, the pathophysiological role of cardiac tissue XO in LV dysfunction remains unclear. We herein investigated the role and functional significance of tissue XO activity in doxorubicin-induced cardiotoxicity. Either doxorubicin (10 mg/kg) or vehicle was intraperitonially administered in a single injection to mice. Mice were treated with or without oral XO-inhibitors (febuxostat 3 mg/kg/day or topiroxostat 5 mg/kg/day) for 8 days starting 24 h before doxorubicin injection. Cardiac tissue XO activity measured by a highly sensitive assay with liquid chromatography/mass spectrometry and cardiac UA content were significantly increased in doxorubicin-treated mice at day 7 and dramatically reduced by XO-inhibitors. Accordingly, XO-inhibitors substantially improved LV ejection fraction (assessed by echocardiography) and LV developed pressure (assessed by ex vivo Langendorff heart perfusion) impaired by doxorubicin administration. This was associated with an increase in XO-derived hydrogen peroxide production with concomitant upregulation of apoptotic and ferroptotic pathways, all of which were reduced by XO-inhibitors. Furthermore, metabolome analyses revealed enhanced purine metabolism in doxorubicin-treated hearts, and XO-inhibitors suppressed the serial metabolic reaction of hypoxanthine-xanthine-UA, the paths of ATP and purine degradation. In summary, doxorubicin administration induces cardiac tissue XO activation associated with impaired LV function. XO-inhibitors attenuate doxorubicin-induced cardiotoxicity through inhibition of XO-derived oxidative stress and cell death signals as well as the maintenance of cardiac energy metabolism associated with modulation of the purine metabolic pathway.
Collapse
Affiliation(s)
- Yoshiro Tanaka
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, Japan
| | - Tomohisa Nagoshi
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, Japan.
| | - Akira Yoshii
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, Japan
| | - Yuhei Oi
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, Japan
| | - Hirotake Takahashi
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, Japan
| | - Haruka Kimura
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, Japan
| | - Keiichi Ito
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, Japan
| | - Yusuke Kashiwagi
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, Japan
| | - Toshikazu D Tanaka
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, Japan
| | - Michihiro Yoshimura
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, Japan
| |
Collapse
|
28
|
Torregroza C, Raupach A, Feige K, Weber NC, Hollmann MW, Huhn R. Perioperative Cardioprotection: General Mechanisms and Pharmacological Approaches. Anesth Analg 2020; 131:1765-1780. [PMID: 33186163 DOI: 10.1213/ane.0000000000005243] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cardioprotection encompasses a variety of strategies protecting the heart against myocardial injury that occurs during and after inadequate blood supply to the heart during myocardial infarction. While restoring reperfusion is crucial for salvaging myocardium from further damage, paradoxically, it itself accounts for additional cell death-a phenomenon named ischemia/reperfusion injury. Therefore, therapeutic strategies are necessary to render the heart protected against myocardial infarction. Ischemic pre- and postconditioning, by short periods of sublethal cardiac ischemia and reperfusion, are still the strongest mechanisms to achieve cardioprotection. However, it is highly impractical and far too invasive for clinical use. Fortunately, it can be mimicked pharmacologically, for example, by volatile anesthetics, noble gases, opioids, propofol, dexmedetomidine, and phosphodiesterase inhibitors. These substances are all routinely used in the clinical setting and seem promising candidates for successful translation of cardioprotection from experimental protocols to clinical trials. This review presents the fundamental mechanisms of conditioning strategies and provides an overview of the most recent and relevant findings on different concepts achieving cardioprotection in the experimental setting, specifically emphasizing pharmacological approaches in the perioperative context.
Collapse
Affiliation(s)
- Carolin Torregroza
- From the Department of Anesthesiology, University Hospital Duesseldorf, Duesseldorf, Germany.,Department of Anesthesiology, Amsterdam University Medical Centers (AUMC), Amsterdam, the Netherlands
| | - Annika Raupach
- From the Department of Anesthesiology, University Hospital Duesseldorf, Duesseldorf, Germany
| | - Katharina Feige
- From the Department of Anesthesiology, University Hospital Duesseldorf, Duesseldorf, Germany
| | - Nina C Weber
- Department of Anesthesiology, Amsterdam University Medical Centers (AUMC), Amsterdam, the Netherlands
| | - Markus W Hollmann
- Department of Anesthesiology, Amsterdam University Medical Centers (AUMC), Amsterdam, the Netherlands
| | - Ragnar Huhn
- From the Department of Anesthesiology, University Hospital Duesseldorf, Duesseldorf, Germany
| |
Collapse
|
29
|
Zhang J, Sun Z, Lin N, Lu W, Huang X, Weng J, Sun S, Zhang C, Yang Q, Zhou G, Guo H, Chi J. Fucoidan from Fucus vesiculosus attenuates doxorubicin-induced acute cardiotoxicity by regulating JAK2/STAT3-mediated apoptosis and autophagy. Biomed Pharmacother 2020; 130:110534. [PMID: 32711244 DOI: 10.1016/j.biopha.2020.110534] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/05/2020] [Accepted: 07/11/2020] [Indexed: 12/21/2022] Open
Abstract
Doxorubicin (DOX) is well-known for its potent antitumor activity but limited by its multiple and serious adverse effects. A major adverse effect is acute cardiotoxicity; yet, its mechanism has not been elucidated. Fucoidan is a multifunctional and nontoxic polysaccharide that is widely studied because of its favorable biological activities and safety. Hence, we proposed that fucoidan may play a protective role in DOX-induced acute cardiotoxicity without causing additional side effects. Sprague-Dawley rats were injected intraperitoneally with a single high dose of DOX to induce acute cardiac injury. Fucoidan was administered orally before DOX injection and AG490, a JAK2 inhibitor, was applied to verify the participation of the JAK2/STAT3 pathway. In vitro, H9C2 cells were treated with the same drugs at different concentrations and intervention times. in vivo and in vitro results demonstrated that DOX administration induced myocardial damage accompanied by acceleratory apoptosis and deficient autophagy in heart tissues or cells, which could be significantly improved by fucoidan supplement. AG490 partly abolished the cardioprotective effects of fucoidan, suggesting the involvement of JAK2 signaling. Additionally, western blotting revealed DOX-induced JAK2/STAT3 pathway activation, which was enhanced by fucoidan and weaken by AG490. Hence, fucoidan exerted a favorable effect on DOX-induced cardiotoxicity by enhancing autophagy and suppressing apoptosis in a JAK2/STAT3-dependent manner, which may provide a promising and novel therapeutic strategy against negative chemotherapy-induced effects.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Cardiology, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, 312000, Zhejiang, China; The First Clinical Medical College, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Zhenzhu Sun
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Na Lin
- Zhejiang Chinese Medical University, Hangzhou, 310000, Zhejiang, China
| | - Wenqiang Lu
- Department of Cardiology, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, 312000, Zhejiang, China
| | - Xingxiao Huang
- Department of Cardiology, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, 312000, Zhejiang, China
| | - Jingfan Weng
- Department of Cardiology, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, 312000, Zhejiang, China
| | - Shimin Sun
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Chuanjing Zhang
- Department of Cardiology, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, 312000, Zhejiang, China
| | - Qi Yang
- Zhejiang Chinese Medical University, Hangzhou, 310000, Zhejiang, China
| | - Guozhong Zhou
- Department of Medical, Shaoxing People's Hospital, Shaoxing, 312000, Zhejiang, China
| | - Hangyuan Guo
- Department of Cardiology, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, 312000, Zhejiang, China; The First Clinical Medical College, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China; Zhejiang Chinese Medical University, Hangzhou, 310000, Zhejiang, China.
| | - Jufang Chi
- Department of Cardiology, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, 312000, Zhejiang, China; The First Clinical Medical College, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China; Zhejiang Chinese Medical University, Hangzhou, 310000, Zhejiang, China.
| |
Collapse
|
30
|
Investigating the Vascular Toxicity Outcomes of the Irreversible Proteasome Inhibitor Carfilzomib. Int J Mol Sci 2020; 21:ijms21155185. [PMID: 32707866 PMCID: PMC7432349 DOI: 10.3390/ijms21155185] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/14/2020] [Accepted: 07/20/2020] [Indexed: 12/19/2022] Open
Abstract
Background: Carfilzomib’s (Cfz) adverse events in myeloma patients include cardiovascular toxicity. Since carfilzomib’s vascular effects are elusive, we investigated the vascular outcomes of carfilzomib and metformin (Met) coadministration. Methods: Mice received: (i) saline; (ii) Cfz; (iii) Met; (iv) Cfz+Met for two consecutive (acute) or six alternate days (subacute protocol). Leucocyte-derived reactive oxygen species (ROS) and serum NOx levels were determined and aortas underwent vascular and molecular analyses. Mechanistic experiments were recapitulated in aged mice who received similar treatment to young animals. Primary murine (prmVSMCs) and aged human aortic smooth muscle cells (HAoSMCs) underwent Cfz, Met and Cfz+Met treatment and viability, metabolic flux and p53-LC3-B expression were measured. Experiments were recapitulated in AngII, CoCl2 and high-glucose stimulated HAoSMCs. Results: Acutely, carfilzomib alone led to vascular hypo-contraction and increased ROS release. Subacutely, carfilzomib increased ROS release without vascular manifestations. Cfz+Met increased PGF2α-vasoconstriction and LC3-B-dependent autophagy in both young and aged mice. In vitro, Cfz+Met led to cytotoxicity and autophagy, while Met and Cfz+Met shifted cellular metabolism. Conclusion: Carfilzomib induces a transient vascular impairment and oxidative burst. Cfz+Met increased vascular contractility and synergistically induced autophagy in all settings. Therefore, carfilzomib cannot be accredited for a permanent vascular dysfunction, while Cfz+Met exert vasoprotective potency.
Collapse
|
31
|
Sala V, Della Sala A, Hirsch E, Ghigo A. Signaling Pathways Underlying Anthracycline Cardiotoxicity. Antioxid Redox Signal 2020; 32:1098-1114. [PMID: 31989842 DOI: 10.1089/ars.2020.8019] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Significance: The cardiac side effects of hematological treatments are a major issue of the growing population of cancer survivors, often affecting patient survival even more than the tumor for which the treatment was initially prescribed. Among the most cardiotoxic drugs are anthracyclines (ANTs), highly potent antitumor agents, which still represent a mainstay in the treatment of hematological and solid tumors. Unfortunately, diagnosis, prevention, and treatment of cardiotoxicity are still unmet clinical needs, which call for a better understanding of the molecular mechanism behind the pathology. Recent Advances: This review article will discuss recent findings on the pathomechanisms underlying the cardiotoxicity of ANTs, spanning from DNA and mitochondrial damage to calcium homeostasis, autophagy, and apoptosis. Special emphasis will be given to the role of reactive oxygen species and their interplay with major signaling pathways. Critical Issues: Although new promising therapeutic targets and new drugs have started to be identified, their efficacy has been mainly proven in preclinical studies and requires clinical validation. Future Directions: Future studies are awaited to confirm the relevance of recently uncovered targets, as well as to identify new druggable pathways, in more clinically relevant models, including, for example, human induced pluripotent stem cell-derived cardiomyocytes.
Collapse
Affiliation(s)
- Valentina Sala
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Angela Della Sala
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| |
Collapse
|
32
|
Zhang H, Tian Y, Liang D, Fu Q, Jia L, Wu D, Zhu X. The Effects of Inhibition of MicroRNA-375 in a Mouse Model of Doxorubicin-Induced Cardiac Toxicity. Med Sci Monit 2020; 26:e920557. [PMID: 32186283 PMCID: PMC7102408 DOI: 10.12659/msm.920557] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Doxorubicin-induced myocardial toxicity is associated with oxidative stress, cardiomyocyte, apoptosis, and loss of contractile function. Previous studies showed that microRNA-375 (miR-375) expression was increased in mouse models of heart failure and clinically, and that inhibition of miR-375 reduced inflammation and increased survival of cardiomyocytes. This study aimed to investigate the effects and mechanisms of inhibition of miR-375 in a mouse model of doxorubicin-induced cardiac toxicity in vivo and in doxorubicin-treated rat and mouse cardiomyocytes in vitro. MATERIAL AND METHODS The mouse model of doxorubicin-induced cardiac toxicity was developed using an intraperitoneal injection of doxorubicin (15 mg/kg diluted in 0.9% saline) for eight days. Treatment was followed by a single subcutaneous injection of miR-375 inhibitor. H9c2 rat cardiac myocytes and adult murine cardiomyocytes (AMCs) were cultured in vitro and treated with doxorubicin, with and without pretreatment with miR-375 inhibitor. RESULTS Doxorubicin significantly upregulated miR-375 expression in vitro and in vivo, and inhibition of miR-375 re-established myocardial redox homeostasis, prevented doxorubicin-induced oxidative stress and cardiomyocyte apoptosis, and activated the PDK1/AKT axis by reducing the direct binding of miR-375 to 3' UTR of the PDK1 gene. Inhibition of PDK1 and AKT abolished the protective role of miR-375 inhibition on doxorubicin-induced oxidative damage. CONCLUSIONS Inhibition of miR-375 prevented oxidative damage in a mouse model of doxorubicin-induced cardiac toxicity in vivo and in doxorubicin-treated rat and mouse cardiomyocytes in vitro through the PDK1/AKT signaling pathway.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Cardiovascular Surgery, Tianjin Medical University General Hospital, Tianjing, China (mainland)
| | - Yikui Tian
- Department of Cardiovascular Surgery, Tianjin Medical University General Hospital, Tianjing, China (mainland)
| | - Degang Liang
- Department of Cardiovascular Surgery, Tianjin Medical University General Hospital, Tianjing, China (mainland)
| | - Qiang Fu
- Department of Cardiovascular Surgery, Tianjin Medical University General Hospital, Tianjing, China (mainland)
| | - Liqun Jia
- Department of Cardiovascular Surgery, Tianjin Medical University General Hospital, Tianjing, China (mainland)
| | - Dawei Wu
- Department of Cardiovascular Surgery, Tianjin Medical University General Hospital, Tianjing, China (mainland)
| | - Xinyuan Zhu
- Department of Cardiovascular Surgery, Tianjin Medical University General Hospital, Tianjing, China (mainland)
| |
Collapse
|
33
|
The Long Noncoding RNA Hotair Regulates Oxidative Stress and Cardiac Myocyte Apoptosis during Ischemia-Reperfusion Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:1645249. [PMID: 32256945 PMCID: PMC7091551 DOI: 10.1155/2020/1645249] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 02/03/2020] [Accepted: 02/17/2020] [Indexed: 12/20/2022]
Abstract
Oxidative stress and subsequent cardiac myocyte apoptosis play central roles in the initiation and progression of myocardial ischemia-reperfusion (I/R) injury. Homeobox transcript antisense intergenic RNA (Hotair) was previously implicated in various heart diseases, yet its role in myocardial I/R injury has not been clearly demonstrated. Mice with cardiac-restricted knockdown or overexpression of Hotair were exposed to I/R surgery. H9c2 cells were cultured and subjected to hypoxia/reoxygenation (H/R) stimulation to further verify the role and underlying mechanisms of Hotair in vitro. Histological examination, molecular detection, and functional parameters were determined in vivo and in vitro. In response to I/R or H/R treatment, Hotair expression was increased in a bromodomain-containing protein 4-dependent manner. Cardiac-restricted knockdown of Hotair exacerbated, whereas Hotair overexpression prevented I/R-induced oxidative stress, cardiac myocyte apoptosis, and cardiac dysfunction. Mechanistically, we observed that Hotair exerted its beneficial effects via activating AMP-activated protein kinase alpha (AMPKα). Further detection revealed that Hotair activated AMPKα through regulating the enhancer of zeste homolog 2/microRNA-451/calcium-binding protein 39 (EZH2/miR-451/Cab39) axis. We provide the evidence that endogenous lncRNA Hotair is an essential negative regulator for oxidative stress and cardiac myocyte apoptosis in myocardial I/R injury, which is dependent on AMPKα activation via the EZH2/miR-451/Cab39 axis.
Collapse
|