51
|
Zhao J, Yu HQ, Ge FQ, Zhang MR, Song YC, Guo DD, Li QH, Zhu H, Hang PZ. 7,8,3'-Trihydroxyflavone prevents doxorubicin-induced cardiotoxicity and mitochondrial dysfunction via activating Akt signaling pathway in H9c2 cells. Cell Signal 2023; 112:110924. [PMID: 37838311 DOI: 10.1016/j.cellsig.2023.110924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/27/2023] [Accepted: 10/09/2023] [Indexed: 10/16/2023]
Abstract
Clinical application of the widely used chemotherapeutic agent, doxorubicin (DOX), is limited by its cardiotoxicity. Mitochondrial dysfunction has been revealed as a crucial factor in DOX-induced cardiotoxicity. 7,8,3'-Trihydroxyflavone (THF) is a mimetic brain-derived neurotrophic factor with neuroprotective effects. However, the potential effects of THF on DOX-induced cardiomyocyte damage and mitochondrial disorders remain unclear. H9c2 cardiomyoblasts were exposed to DOX and/or THF at different concentrations. Cardiomyocyte injury was evaluated using lactate dehydrogenase (LDH) assay and Live/Dead cytotoxicity kit. Meanwhile, mitochondrial membrane potential (MMP), morphology, mitochondrial reactive oxygen species (mito-ROS) production, and the oxygen consumption rate of cardiomyocytes were measured. The protein levels of key mitochondria-related factors such as adenosine monophosphate-activated protein kinase (AMPK), mitofusin 2 (Mfn2), dynamin-related protein 1 (Drp1), and optic atrophy protein 1 (OPA1) were examined. We found that THF reduced LDH content and death ratio of DOX-treated cardiomyocytes in a concentration-dependent manner, while increasing MMP without significantly affecting the routine and maximum capacity of mitochondrial respiration. Mechanistically, THF increased the activity of Akt and protein levels of Mfn2 and heme oxygenase 1 (HO-1). Moreover, inhibition of Akt reversed the protective role of THF, increased mito-ROS levels, and repressed Mfn2 and HO-1 expression. Therefore, we conclude, THF relieves DOX-induced cardiotoxicity and improves mitochondrial function by activating Akt-mediated Mfn2 and HO-1 pathways. This finding provides promising therapeutic insights for DOX-induced cardiac dysfunction.
Collapse
Affiliation(s)
- Jing Zhao
- Department of Pharmacy, Clinical Medical College, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou 225001, China
| | - Hua-Qing Yu
- Department of Pharmacy, Clinical Medical College, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou 225001, China; College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Feng-Qin Ge
- Department of Pharmacy, Clinical Medical College, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou 225001, China; Medical College, Yangzhou University, Yangzhou 225009, China
| | - Man-Ru Zhang
- Department of Pharmacy, Clinical Medical College, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou 225001, China; College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Yu-Chen Song
- Department of Pharmacy, Clinical Medical College, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou 225001, China; Medical College, Yangzhou University, Yangzhou 225009, China
| | - Dan-Dan Guo
- Department of Pharmacy, Clinical Medical College, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou 225001, China; Medical College, Yangzhou University, Yangzhou 225009, China
| | - Qi-Hang Li
- Department of Pharmacy, Clinical Medical College, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou 225001, China; Medical College, Yangzhou University, Yangzhou 225009, China
| | - Hua Zhu
- Department of Pharmacy, Clinical Medical College, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou 225001, China.
| | - Peng-Zhou Hang
- Department of Pharmacy, Clinical Medical College, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou 225001, China.
| |
Collapse
|
52
|
Fang G, Li X, Yang F, Huang T, Qiu C, Peng K, Yang Y, Lan C. Galangin attenuates doxorubicin-induced cardiotoxicity via activating nuclear factor erythroid 2-related factor 2/heme oxygenase 1 signaling pathway to suppress oxidative stress and inflammation. Phytother Res 2023; 37:5854-5870. [PMID: 37655750 DOI: 10.1002/ptr.7991] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/07/2023] [Accepted: 08/10/2023] [Indexed: 09/02/2023]
Abstract
Doxorubicin (DOX) has aroused contradiction between its potent anti-tumor capacity and severe cardiotoxicity. Galangin (Gal) possesses antioxidant, anti-inflammatory, and antiapoptotic activities. We aimed to explore the role and underlying mechanisms of Gal on DOX-induced cardiotoxicity. Mice were intraperitoneally injected with DOX (3 mg/kg, every 2 days for 2 weeks) to generate cardiotoxicity model and Gal (15 mg/kg, 2 weeks) was co-administered via gavage daily. Nuclear factor erythroid 2-related factor 2 (Nrf2) specific inhibitor, ML385, was employed to explore the underlying mechanisms. Compared to DOX-insulted mice, Gal effectively improved cardiac dysfunction and ameliorated myocardial damage. DOX-induced increase of reactive oxygen species, malondialdehyde, and NADPH oxidase activity and downregulation of superoxide dismutase (SOD) activity were blunted by Gal. Gal also markedly blocked increase of IL-1β, IL-6, and TNF-α in DOX-insulted heart. Mechanistically, Gal reversed DOX-induced downregulation of Nrf2, HO-1, and promoted nuclear translocation of Nrf2. ML385 markedly blunted the cardioprotective effects of Gal, as well as inhibitive effects on oxidative stress and inflammation. Gal ameliorates DOX-induced cardiotoxicity by suppressing oxidative stress and inflammation via activating Nrf2/HO-1 signaling pathway. Gal may serve as a promising cardioprotective agent for DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Guangyao Fang
- Department of Cardiology, General Hospital of Western Theater Command, Chengdu, China
- College of Medicine, Southwest Jiaotong University, Chengdu, China
| | - Xiuchuan Li
- Department of Cardiology, General Hospital of Western Theater Command, Chengdu, China
| | - Fengyuan Yang
- Department of Nephrology, General Hospital of Western Theater Command, Chengdu, China
| | - Ting Huang
- Department of Medical Oncology, People's Hospital of Luotian County, Huanggang, China
| | - Chenming Qiu
- Department of Burn and Plastic Surgery, General Hospital of Western Theater Command, Chengdu, China
| | - Ke Peng
- Department of Cardiology, General Hospital of Western Theater Command, Chengdu, China
| | - Yongjian Yang
- Department of Cardiology, General Hospital of Western Theater Command, Chengdu, China
- College of Medicine, Southwest Jiaotong University, Chengdu, China
| | - Cong Lan
- Department of Cardiology, General Hospital of Western Theater Command, Chengdu, China
| |
Collapse
|
53
|
Wu Y, Zhao J, Tian Y, Jin H. Cellular functions of heat shock protein 20 (HSPB6) in cancer: A review. Cell Signal 2023; 112:110928. [PMID: 37844714 DOI: 10.1016/j.cellsig.2023.110928] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/07/2023] [Accepted: 10/13/2023] [Indexed: 10/18/2023]
Abstract
Heat shock proteins (HSP) are a large family of peptide proteins that are widely found in cells. Studies have shown that the expression and function of HSPs in cells are very complex, and they can participate in cellular physiological and pathological processes through multiple pathways. Multiple heat shock proteins are associated with cancer cell growth, proliferation, metastasis, and resistance to anticancer drugs, and they play a key role in cancer development by ensuring the correct folding or degradation of proteins in cancer cells. As research hotspots, HSP90, HSP70 and HSP27 have been extensively studied in cancer so far. However, HSP20, also referred to as HSPB6, as a member of the small heat shock protein family, has been shown to play an important role in the cardiovascular system, but little research has been conducted on HSP20 in cancer. This review summarizes the current cellular functions of HSP20 in different cancer types, as well as its effects on cancer proliferation, progression, prognosis, and its other functions in cancer, to illustrate the close association between HSP20 and cancer. We show that, unlike most HSPs, HSP20 mainly plays an active anticancer role in cancer development, which is expected to provide new ideas and help for cancer diagnosis and treatment and research.
Collapse
Affiliation(s)
- Yifeng Wu
- Department of General Surgery, Wuxi 9th People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu 214000, People's Republic of China
| | - Jinjin Zhao
- Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, People's Republic of China
| | - Yun Tian
- Department of Oncology, Jiangsu Province Hospital of Chinese Medicine Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, People's Republic of China.
| | - Hongdou Jin
- Department of General Surgery, Wuxi 9th People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu 214000, People's Republic of China.
| |
Collapse
|
54
|
Tai P, Chen X, Jia G, Chen G, Gong L, Cheng Y, Li Z, Wang H, Chen A, Zhang G, Zhu Y, Xiao M, Wang Z, Liu Y, Shan D, He D, Li M, Zhan T, Khan A, Li X, Zeng X, Li C, Ouyang D, Ai K, Chen X, Liu D, Liu Z, Wei D, Cao K. WGX50 mitigates doxorubicin-induced cardiotoxicity through inhibition of mitochondrial ROS and ferroptosis. J Transl Med 2023; 21:823. [PMID: 37978379 PMCID: PMC10655295 DOI: 10.1186/s12967-023-04715-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 11/09/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND Doxorubicin (DOX)-induced cardiotoxicity (DIC) is a major impediment to its clinical application. It is indispensable to explore alternative treatment molecules or drugs for mitigating DIC. WGX50, an organic extract derived from Zanthoxylum bungeanum Maxim, has anti-inflammatory and antioxidant biological activity, however, its function and mechanism in DIC remain unclear. METHODS We established DOX-induced cardiotoxicity models both in vitro and in vivo. Echocardiography and histological analyses were used to determine the severity of cardiac injury in mice. The myocardial damage markers cTnT, CK-MB, ANP, BNP, and ferroptosis associated indicators Fe2+, MDA, and GPX4 were measured using ELISA, RT-qPCR, and western blot assays. The morphology of mitochondria was investigated with a transmission electron microscope. The levels of mitochondrial membrane potential, mitochondrial ROS, and lipid ROS were detected using JC-1, MitoSOX™, and C11-BODIPY 581/591 probes. RESULTS Our findings demonstrate that WGX50 protects DOX-induced cardiotoxicity via restraining mitochondrial ROS and ferroptosis. In vivo, WGX50 effectively relieves doxorubicin-induced cardiac dysfunction, cardiac injury, fibrosis, mitochondrial damage, and redox imbalance. In vitro, WGX50 preserves mitochondrial function by reducing the level of mitochondrial membrane potential and increasing mitochondrial ATP production. Furthermore, WGX50 reduces iron accumulation and mitochondrial ROS, increases GPX4 expression, and regulates lipid metabolism to inhibit DOX-induced ferroptosis. CONCLUSION Taken together, WGX50 protects DOX-induced cardiotoxicity via mitochondrial ROS and the ferroptosis pathway, which provides novel insights for WGX50 as a promising drug candidate for cardioprotection.
Collapse
Affiliation(s)
- Panpan Tai
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Xinyu Chen
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Guihua Jia
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Guanjun Chen
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Lian Gong
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Yaxin Cheng
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhuan Li
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha, 410013, China
- The Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Hunan Normal University School of Medicine, Changsha, 410013, China
- Department of Pharmacy, Hunan Normal University School of Medicine, Changsha, 410013, China
| | - Heng Wang
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Aiyan Chen
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Ganghua Zhang
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Yuxing Zhu
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Mengqing Xiao
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhanwang Wang
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Yunqing Liu
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Dongyong Shan
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Dong He
- Staff Hospital of Central South University, Central South University, Changsha, China
| | - Moying Li
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Tianzuo Zhan
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Abbas Khan
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaohui Li
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Xiangxiang Zeng
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, China
| | - Chaopeng Li
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha Duxact Biotech Co., Ltd, Changsha, China
| | - Dongsheng Ouyang
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha Duxact Biotech Co., Ltd, Changsha, China
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
| | - Kelong Ai
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Xuan Chen
- College of Horticulture, Hunan Agricultural University, Changsha, China
- State Key Laboratory of Subhealth Intervention Technology, Changsha, China
- National Research Center of Engineering Technology for Utilization Ingredients From Botanicals, Changsha, China
| | - Dongbo Liu
- College of Horticulture, Hunan Agricultural University, Changsha, China
- State Key Laboratory of Subhealth Intervention Technology, Changsha, China
- National Research Center of Engineering Technology for Utilization Ingredients From Botanicals, Changsha, China
| | - Zhonghua Liu
- National Research Center of Engineering Technology for Utilization Ingredients From Botanicals, Changsha, China
| | - Dongqing Wei
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China.
| | - Ke Cao
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
55
|
Durço AO, Souza DS, Rhana P, Costa AD, Marques LP, Santos LABO, de Souza Araujo AA, de Aragão Batista MV, Roman-Campos D, Santos MRVD. d-Limonene complexed with cyclodextrin attenuates cardiac arrhythmias in an experimental model of doxorubicin-induced cardiotoxicity: Possible involvement of calcium/calmodulin-dependent protein kinase type II. Toxicol Appl Pharmacol 2023; 474:116609. [PMID: 37392997 DOI: 10.1016/j.taap.2023.116609] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 06/15/2023] [Accepted: 06/26/2023] [Indexed: 07/03/2023]
Abstract
BACKGROUND Arrhythmias are one manifestation of the cardiotoxicity that results from doxorubicin (Doxo) administration. Although cardiotoxicity is an anticipated outcome in anticancer therapies, there is still a lack of treatment options available for its effective management. This study sought to evaluate the possible cardioprotective effect of complex d-limonene (DL) plus hydroxypropyl-β-cyclodextrin (HβDL) during treatment with Doxo, focusing on the arrhythmic feature. METHODS Cardiotoxicity was induced in Swiss mice with Doxo 20 mg/kg, with 10 mg/kg of HβDL being administered 30 min before the Doxo. Plasma CK-MB and LDH levels were analyzed. Cellular excitability and susceptibility to cardiac and cardiomyocyte arrhythmias were evaluated using in vivo (pharmacological cardiac stress) and in vitro (burst pacing) ECG protocols. Ca2+ dynamics were also investigated. The expression of CaMKII and its activation by phosphorylation and oxidation were evaluated by western blot, and molecular docking was used to analyze the possible interaction between DL and CaMKII. RESULTS Electrocardiograms showed that administration of 10 mg/kg of HβDL prevented Doxo-induced widening of the QRS complex and QT interval. HβDL also prevented cardiomyocyte electrophysiological changes that trigger cellular arrhythmias, such as increases in action potential duration and variability; decreased the occurrence of delayed afterdepolarizations (DADs) and triggered activities (TAs), and reduced the incidence of arrhythmia in vivo. Ca2+ waves and CaMKII overactivation caused by phosphorylation and oxidation were also decreased. In the in silico study, DL showed potential inhibitory interaction with CaMKII. CONCLUSION Our results show that 10 mg/kg of βDL protects the heart against Doxo-induced cardiotoxicity arrhythmias, and that this is probably due to its inhibitory effect on CaMKII hyperactivation.
Collapse
Affiliation(s)
- Aimée Obolari Durço
- Health Science Graduate Program, Federal University of Sergipe, Aracaju, Brazil
| | - Diego Santos Souza
- Department of Biophysics, Federal University of São Paulo, São Paulo, Brazil
| | - Paula Rhana
- Department of Physiology and Membrane Biology, University of California, Davis, USA
| | | | | | | | - Adriano Antunes de Souza Araujo
- Health Science Graduate Program, Federal University of Sergipe, Aracaju, Brazil; Department of Pharmacy, Federal University of Sergipe, São Cristóvão, Brazil
| | | | - Danilo Roman-Campos
- Department of Biophysics, Federal University of São Paulo, São Paulo, Brazil
| | - Marcio Roberto Viana Dos Santos
- Health Science Graduate Program, Federal University of Sergipe, Aracaju, Brazil; Department of Physiology, Federal University of Sergipe, São Cristóvão, Brazil.
| |
Collapse
|
56
|
Airo AA, Sunela KL. Late complications and survival of soft tissue sarcoma patients after adjuvant chemotherapy - a single tertiary centre experience. Acta Oncol 2023; 62:1152-1156. [PMID: 37571925 DOI: 10.1080/0284186x.2023.2245551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 07/27/2023] [Indexed: 08/13/2023]
|
57
|
Fazio A, Evangelisti C, Cappellini A, Mongiorgi S, Koufi FD, Neri I, Marvi MV, Russo M, Ghigo A, Manzoli L, Fiume R, Ratti S. Emerging Roles of Phospholipase C Beta Isozymes as Potential Biomarkers in Cardiac Disorders. Int J Mol Sci 2023; 24:13096. [PMID: 37685903 PMCID: PMC10487445 DOI: 10.3390/ijms241713096] [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: 08/03/2023] [Revised: 08/19/2023] [Accepted: 08/21/2023] [Indexed: 09/10/2023] Open
Abstract
Phospholipase C (PLC) enzymes represent crucial participants in the plasma membrane of mammalian cells, including the cardiac sarcolemmal (SL) membrane of cardiomyocytes. They are responsible for the hydrolysis of phosphatidylinositol 4,5-bisphosphate (PtdIns(4,5)P2) into 1,2-diacylglycerol (DAG) and inositol (1,4,5) trisphosphate (Ins(1,4,5)P3), both essential lipid mediators. These second messengers regulate the intracellular calcium (Ca2+) concentration, which activates signal transduction cascades involved in the regulation of cardiomyocyte activity. Of note, emerging evidence suggests that changes in cardiomyocytes' phospholipid profiles are associated with an increased occurrence of cardiovascular diseases, but the underlying mechanisms are still poorly understood. This review aims to provide a comprehensive overview of the significant impact of PLC on the cardiovascular system, encompassing both physiological and pathological conditions. Specifically, it focuses on the relevance of PLCβ isoforms as potential cardiac biomarkers, due to their implications for pathological disorders, such as cardiac hypertrophy, diabetic cardiomyopathy, and myocardial ischemia/reperfusion injury. Gaining a deeper understanding of the mechanisms underlying PLCβ activation and regulation is crucial for unraveling the complex signaling networks involved in healthy and diseased myocardium. Ultimately, this knowledge holds significant promise for advancing the development of potential therapeutic strategies that can effectively target and address cardiac disorders by focusing on the PLCβ subfamily.
Collapse
Affiliation(s)
- Antonietta Fazio
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (A.F.); (C.E.); (A.C.); (S.M.); (F.-D.K.); (I.N.); (M.V.M.); (L.M.)
| | - Camilla Evangelisti
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (A.F.); (C.E.); (A.C.); (S.M.); (F.-D.K.); (I.N.); (M.V.M.); (L.M.)
| | - Alessandra Cappellini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (A.F.); (C.E.); (A.C.); (S.M.); (F.-D.K.); (I.N.); (M.V.M.); (L.M.)
| | - Sara Mongiorgi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (A.F.); (C.E.); (A.C.); (S.M.); (F.-D.K.); (I.N.); (M.V.M.); (L.M.)
| | - Foteini-Dionysia Koufi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (A.F.); (C.E.); (A.C.); (S.M.); (F.-D.K.); (I.N.); (M.V.M.); (L.M.)
| | - Irene Neri
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (A.F.); (C.E.); (A.C.); (S.M.); (F.-D.K.); (I.N.); (M.V.M.); (L.M.)
| | - Maria Vittoria Marvi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (A.F.); (C.E.); (A.C.); (S.M.); (F.-D.K.); (I.N.); (M.V.M.); (L.M.)
| | - Michele Russo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center “Guido Tarone”, University of Torino, 10126 Torino, Italy; (M.R.); (A.G.)
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center “Guido Tarone”, University of Torino, 10126 Torino, Italy; (M.R.); (A.G.)
| | - Lucia Manzoli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (A.F.); (C.E.); (A.C.); (S.M.); (F.-D.K.); (I.N.); (M.V.M.); (L.M.)
| | - Roberta Fiume
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (A.F.); (C.E.); (A.C.); (S.M.); (F.-D.K.); (I.N.); (M.V.M.); (L.M.)
| | - Stefano Ratti
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (A.F.); (C.E.); (A.C.); (S.M.); (F.-D.K.); (I.N.); (M.V.M.); (L.M.)
| |
Collapse
|
58
|
Mazevet M, Belhadef A, Ribeiro M, Dayde D, Llach A, Laudette M, Belleville T, Mateo P, Gressette M, Lefebvre F, Chen J, Bachelot-Loza C, Rucker-Martin C, Lezoualch F, Crozatier B, Benitah JP, Vozenin MC, Fischmeister R, Gomez AM, Lemaire C, Morel E. EPAC1 inhibition protects the heart from doxorubicin-induced toxicity. eLife 2023; 12:e83831. [PMID: 37551870 PMCID: PMC10484526 DOI: 10.7554/elife.83831] [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: 09/29/2022] [Accepted: 08/03/2023] [Indexed: 08/09/2023] Open
Abstract
Anthracyclines, such as doxorubicin (Dox), are widely used chemotherapeutic agents for the treatment of solid tumors and hematologic malignancies. However, they frequently induce cardiotoxicity leading to dilated cardiomyopathy and heart failure. This study sought to investigate the role of the exchange protein directly activated by cAMP (EPAC) in Dox-induced cardiotoxicity and the potential cardioprotective effects of EPAC inhibition. We show that Dox induces DNA damage and cardiomyocyte cell death with apoptotic features. Dox also led to an increase in both cAMP concentration and EPAC1 activity. The pharmacological inhibition of EPAC1 (with CE3F4) but not EPAC2 alleviated the whole Dox-induced pattern of alterations. When administered in vivo, Dox-treated WT mice developed a dilated cardiomyopathy which was totally prevented in EPAC1 knock-out (KO) mice. Moreover, EPAC1 inhibition potentiated Dox-induced cell death in several human cancer cell lines. Thus, EPAC1 inhibition appears as a potential therapeutic strategy to limit Dox-induced cardiomyopathy without interfering with its antitumoral activity.
Collapse
Affiliation(s)
| | | | | | | | | | - Marion Laudette
- Institut des Maladies Metaboliques et Cardiovasculaires - I2MC, INSERM, Université de ToulouseToulouseFrance
| | - Tiphaine Belleville
- Innovations Thérapeutiques en Hémostase - UMR-S 1140, INSERM, Faculté de Pharmacie, Université Paris Descartes, Sorbonne Paris CitéParisFrance
| | | | | | | | - Ju Chen
- Basic Cardiac Research UCSD School of Medicine La JollaSan DiegoUnited States
| | - Christilla Bachelot-Loza
- Innovations Thérapeutiques en Hémostase - UMR-S 1140, INSERM, Faculté de Pharmacie, Université Paris Descartes, Sorbonne Paris CitéParisFrance
| | - Catherine Rucker-Martin
- Faculté de Médecine, Université Paris-SaclayLe Kremlin BicêtreFrance
- Inserm UMR_S 999, Hôpital Marie LannelongueLe Plessis RobinsonFrance
| | - Frank Lezoualch
- Institut des Maladies Metaboliques et Cardiovasculaires - I2MC, INSERM, Université de ToulouseToulouseFrance
| | | | | | | | | | | | - Christophe Lemaire
- Université Paris-SaclayOrsayFrance
- Université Paris-Saclay, UVSQ, InsermOrsayFrance
| | | |
Collapse
|
59
|
Ghigo A, Ameri P. p53 at the Intersection of Cardiac Fibroblast Proliferation and Activation: Answers and Questions. Circ Res 2023; 133:288-290. [PMID: 37471487 DOI: 10.1161/circresaha.123.323209] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/22/2023]
Affiliation(s)
- Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone," University of Torino, Italy (A.G.)
| | - Pietro Ameri
- Department of Internal Medicine, University of Genova, Italy (P.A.)
- Cardiovascular Disease Unit, Cardiac, Thoracic and Vascular Department, IRCCS Ospedale Policlinico San Martino - IRCCS Italian Cardiology Network, Genova, Italy (P.A.)
| |
Collapse
|
60
|
Ziaei E, de Paiva IM, Yao SJ, Sarrami N, Mehinrad P, Lai J, Lavasanifar A, Kaur K. Peptide-Drug Conjugate Targeting Keratin 1 Inhibits Triple-Negative Breast Cancer in Mice. Mol Pharm 2023; 20:3570-3577. [PMID: 37307328 PMCID: PMC10699791 DOI: 10.1021/acs.molpharmaceut.3c00189] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Selective delivery of chemotherapy to the tumor site while sparing healthy cells and tissues is an attractive approach for cancer treatment. Carriers such as peptides can facilitate selective tumor targeting and payload delivery. Peptides with specific affinity for the overexpressed cell-surface receptors in cancer cells are conjugated to chemotherapy to afford peptide-drug conjugates (PDCs) that show selective uptake by cancer cells. Using a 10-mer linear peptide (WxEAAYQrFL) called 18-4 that targets and binds breast cancer cells, we designed a peptide 18-4-doxorubicin (Dox) conjugate with high specific toxicity toward triple-negative breast cancer (TNBC) MDA-MB-231 cells and 30-fold lower toxicity to normal breast MCF10A epithelial cells. Here, we elucidate the in vivo activity of this potent and tumor-selective peptide 18-4-Dox conjugate in mice bearing orthotopic MDA-MB-231 tumors. Mice treated with four weekly injections of the conjugate showed significantly lower tumor volumes compared to mice treated with free Dox at an equivalent Dox dose. Immunohistochemical (IHC) analysis of mice tissues revealed that treatment with a low dose of PDC (2.5 mg/kg of Dox equiv) reduced the expression of proliferation markers (PCNA and Ki-67) and increased apoptosis (evidenced by increased caspase-3 expression). At the same dose of free Dox (2.5 mg/kg), the expression of these markers was similar to that of saline treatment. Accordingly, significantly more Dox accumulated in tumors of conjugate-treated mice (7-fold) compared to the Dox-treated mice, while lower levels of Dox were observed in the liver, heart, and lungs of peptide-Dox conjugate-treated mice (up to 3-fold less) than Dox-treated mice. The IHC analysis of keratin 1 (K1), the receptor for peptide 18-4, revealed K1 upregulation in tumors and low levels in normal mammary fat pad and liver tissues from mice, suggesting preferential uptake of PDCs by TNBC to be K1 receptor-mediated. Taken together, our data support the use of a PDC approach to deliver chemotherapy selectively to the TNBC to inhibit tumor growth.
Collapse
Affiliation(s)
- Elmira Ziaei
- Chapman University School of Pharmacy (CUSP), Harry and Diane Rinker Health Science Campus, Chapman University, Irvine, CA, 92618-1908, USA
| | - Igor Moura de Paiva
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, T6G 2E1, Canada
| | - Shih-Jing Yao
- Chapman University School of Pharmacy (CUSP), Harry and Diane Rinker Health Science Campus, Chapman University, Irvine, CA, 92618-1908, USA
| | - Nasim Sarrami
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, T6G 2E1, Canada
| | - Parnian Mehinrad
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, T6G 2E1, Canada
| | - Justine Lai
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, T6G 2E1, Canada
| | - Afsaneh Lavasanifar
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, T6G 2E1, Canada
| | - Kamaljit Kaur
- Chapman University School of Pharmacy (CUSP), Harry and Diane Rinker Health Science Campus, Chapman University, Irvine, CA, 92618-1908, USA
| |
Collapse
|
61
|
Zhao J, Zhang N, Ma X, Li M, Feng H. The dual role of ferroptosis in anthracycline-based chemotherapy includes reducing resistance and increasing toxicity. Cell Death Discov 2023; 9:184. [PMID: 37344500 DOI: 10.1038/s41420-023-01483-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 06/02/2023] [Accepted: 06/14/2023] [Indexed: 06/23/2023] Open
Abstract
In conjunction with previous studies, we have noted that ferroptosis, as an emerging mode of regulated cell death (RCD), is intimately related to anthracycline pharmacotherapy. Not only does ferroptosis significantly modulate tumour resistance and drug toxicity, which are core links of the relevant chemotherapeutic process, but it also appears to play a conflicting role that has yet to be appreciated. By targeting the dual role of ferroptosis in anthracycline-based chemotherapy, this review aims to focus on the latest findings at this stage, identify the potential associations and provide novel perspectives for subsequent research directions and therapeutic strategies.
Collapse
Affiliation(s)
- Jiazheng Zhao
- Department of Orthopedics, The Fourth Hospital of Hebei Medical University, 12 Health Road, Shijiazhuang, Hebei, 050011, China
| | - Ning Zhang
- Department of Cardiology, The Fourth Hospital of Hebei Medical University, 12 Health Road, Shijiazhuang, Hebei, 050011, China
| | - Xiaowei Ma
- Departments of Orthopedics, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Nanli, Panjiayuan, Chaoyang District, Beijing, 100021, China
| | - Ming Li
- Department of Orthopedics, The Second Hospital of Hebei Medical University, 215 Heping Road, Shijia-zhuang, Hebei, China
| | - Helin Feng
- Departments of Orthopedics, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Nanli, Panjiayuan, Chaoyang District, Beijing, 100021, China.
| |
Collapse
|
62
|
Chen M, Xue J, Wang M, Yang J, Chen T. Cardiovascular Complications of Pan-Cancer Therapies: The Need for Cardio-Oncology. Cancers (Basel) 2023; 15:cancers15113055. [PMID: 37297017 DOI: 10.3390/cancers15113055] [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: 04/12/2023] [Revised: 05/28/2023] [Accepted: 06/01/2023] [Indexed: 06/12/2023] Open
Abstract
It is more likely that a long-term survivor will have both cardiovascular disease and cancer on account of the progress in cancer therapy. Cardiotoxicity is a well-recognized and highly concerning adverse effect of cancer therapies. This side effect can manifest in a proportion of cancer patients and may lead to the discontinuation of potentially life-saving anticancer treatment regimens. Consequently, this discontinuation may adversely affect the patient's survival prognosis. There are various underlying mechanisms by which each anticancer treatment affects the cardiovascular system. Similarly, the incidence of cardiovascular events varies with different protocols for malignant tumors. In the future, comprehensive cardiovascular risk assessment and clinical monitoring should be considered for cancer treatments. Baseline cardiovascular evaluation risk should be emphasized prior to initiating clinical therapy in patients. Additionally, we highlight that there is a need for cardio-oncology to avoid or prevent cardiovascular side effects. Cardio-oncology service is based on identifying cardiotoxicity, developing strategies to reduce these toxicities, and minimizing long-term cardiotoxic effects.
Collapse
Affiliation(s)
- Mengjia Chen
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Jianing Xue
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Maoling Wang
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Junyao Yang
- Department of Laboratory Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Ting Chen
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
- Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 310058, China
| |
Collapse
|
63
|
Grover SP, Bharathi V, Posma JJ, Griffin JH, Palumbo JS, Mackman N, Antoniak S. Thrombin-mediated activation of PAR1 enhances doxorubicin-induced cardiac injury in mice. Blood Adv 2023; 7:1945-1953. [PMID: 36477178 PMCID: PMC10189413 DOI: 10.1182/bloodadvances.2022008637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 11/21/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022] Open
Abstract
The chemotherapeutic drug doxorubicin is cardiotoxic and can cause irreversible heart failure. In addition to being cardiotoxic, doxorubicin also induces the activation of coagulation. We determined the effect of thrombin-mediated activation of protease-activated receptor 1 (PAR1) on doxorubicin-induced cardiac injury. Administration of doxorubicin to mice resulted in a significant increase in plasma prothrombin fragment 1+2, thrombin-antithrombin complexes, and extracellular vesicle tissue factor activity. Doxorubicin-treated mice expressing low levels of tissue factor, but not factor XII-deficient mice, had reduced plasma thrombin-antithrombin complexes compared to controls. To evaluate the role of thrombin-mediated activation of PAR1, transgenic mice insensitive to thrombin (Par1R41Q) or activated protein C (Par1R46Q) were subjected to acute and chronic models of doxorubicin-induced cardiac injury and compared with Par1 wild-type (Par1+/+) and PAR1 deficient (Par1-/-) mice. Par1R41Q and Par1-/- mice, but not Par1R46Q mice, demonstrated similar reductions in the cardiac injury marker cardiac troponin I, preserved cardiac function, and reduced cardiac fibrosis compared to Par1+/+ controls after administration of doxorubicin. Furthermore, inhibition of Gαq signaling downstream of PAR1 with the small molecule inhibitor Q94 significantly preserved cardiac function in Par1+/+ mice, but not in Par1R41Q mice subjected to the acute model of cardiac injury when compared to vehicle controls. In addition, mice with PAR1 deleted in either cardiomyocytes or cardiac fibroblasts demonstrated reduced cardiac injury compared to controls. Taken together, these data suggest that thrombin-mediated activation of PAR1 contributes to doxorubicin-induced cardiac injury.
Collapse
Affiliation(s)
- Steven P. Grover
- University of North Carolina (UNC) Blood Research Center, Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Vanthana Bharathi
- University of North Carolina (UNC) Blood Research Center, Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Jens J. Posma
- University of North Carolina (UNC) Blood Research Center, Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Laboratory for Clinical Thrombosis and Haemostasis, Department of Internal Medicine, Cardiovascular Research Institute, Maastricht University Medical Center, Maastricht, The Netherlands
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - John H. Griffin
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
- Department of Medicine, University of California San Diego, San Diego, CA
| | - Joseph S. Palumbo
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH
| | - Nigel Mackman
- University of North Carolina (UNC) Blood Research Center, Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Silvio Antoniak
- UNC Blood Research Center, UNC Lineberger Comprehensive Cancer Center, Department of Pathology and Laboratory Medicine, UNC McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC
| |
Collapse
|
64
|
Chen L, Qin Z, Ruan ZB. Hyperoside alleviates doxorubicin-induced myocardial cells apoptosis by inhibiting the apoptosis signal-regulating kinase 1/p38 pathway. PeerJ 2023; 11:e15315. [PMID: 37220525 PMCID: PMC10200097 DOI: 10.7717/peerj.15315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 04/06/2023] [Indexed: 05/25/2023] Open
Abstract
Background Cardiotoxicity is a side effect of the anthracycline broad-spectrum anti-tumor agent, doxorubicin (DOX). Hyperoside, a flavonoid glycoside extracted from many herbs, has anti-apoptotic and anticancer properties. However, its impact on the alleviation of DOX-induced apoptosis in cardiomyocytes remains elusive. Methods The HL-1 cell line was treated with 100 µ M hyperoside for 1 h prior to treatment with 100 µ M hyperoside and 1 µ M DOX for 24 h. The cell counting kit-8 (CCK-8) assay was used to detect cell viability; DCFH-DA fluorescent probe was used to detect (reactive oxygen species) ROS; biochemical methods were used to detect the activity of glutathione (GSH), catalase (CAT), superoxide dismutase (SOD), malondialdehyde (MDA); the degree of apoptosis following DOX insult was assessed using immunofluorescence staining and terminal deoxynucleotidyl transferase mediated deoxy uridine triphosphate nick end labeling (TUNEL) assay; the change in protein expression of apoptosis signal-regulating kinase 1 (ASK1), p38, and apoptosis markers was determined using western blot. Results Hyperoside ameliorated DOX-induced oxidative stress in HL-1 cells, up-regulated GSH, SOD and CAT activity, reduced ROS production and inhibited MDA overproduction. Moreover, in addition to promoting HL-1 cell apoptosis, DOX administration also increased B-cell lymphoma (Bcl)-2-associated X-protein and cleaved caspase-3 protein levels and decreased Bcl-2 protein level. Hyperoside therapy, however, significantly reversed the impact of DOX on the cardiomyocytes. Mechanically, DOX treatment increased the phosphorylation of the ASK1/p38 axis whereas hyperoside treatment attenuated those changes. In a further step, hyperoside synergizes with DOX to kill MDA-MB-231 cells. Conclusions Hyperoside protects HL-1 cells from DOX-induced cardiotoxicity by inhibiting the ASK1/p38 signaling pathway. Meanwhile, hyperoside maintained the cytotoxicity of DOX in MDA-MB-231 cells.
Collapse
Affiliation(s)
- Lingxia Chen
- Department of Cardiology, Nanjing University of Chinese Medicine, Nanjing, China
- Department of Cardiology, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, China
| | - Zhi Qin
- Department of Cardiology, Nanjing University of Chinese Medicine, Nanjing, China
- Department of Cardiology, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, China
| | - Zhong-bao Ruan
- Department of Cardiology, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, China
| |
Collapse
|
65
|
Ghigo A, Meijers WC, Rhee JW, Ameri P. Editorial: Cardio-oncology and reverse cardio-oncology: the manifold interconnections between heart failure and cancer. Front Physiol 2023; 14:1205810. [PMID: 37228811 PMCID: PMC10203590 DOI: 10.3389/fphys.2023.1205810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 04/28/2023] [Indexed: 05/27/2023] Open
Affiliation(s)
- Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center “Guido Tarone”, University of Torino, Torino, Italy
| | | | - June-Wha Rhee
- Department of Medicine, City of Hope National Medical Center, Duarte, CA, United States
| | - Pietro Ameri
- Department of Internal Medicine, University of Genova, Genova, Italy
- Cardiovascular Disease Unit, Cardiac, Thoracic and Vascular Department, IRCCS Ospedale Policlinico San Martino—IRCCS Italian Cardiology Network, Genova, Italy
| |
Collapse
|
66
|
Chen J, Cheng C, Fan L, Xu X, Chen J, Feng Y, Tang Y, Yang C. Assessment of left heart dysfunction to predict doxorubicin cardiotoxicity in children with lymphoma. Front Pediatr 2023; 11:1163664. [PMID: 37215605 PMCID: PMC10196234 DOI: 10.3389/fped.2023.1163664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 04/11/2023] [Indexed: 05/24/2023] Open
Abstract
Objectives The objectives of this study were to assess the changes in the left myocardial function after chemotherapy for childhood lymphoma and observe the predictive or monitor value for cancer treatment-related cardiac dysfunction (CTRCD) by speckle-tracking echocardiography. Methods A total of 23 children with histopathological diagnoses of lymphoma were included, with age-matched normal controls. Comparative analysis of clinical serological tests and left heart strain parameters in children with lymphoma, including left ventricular global longitudinal strain (LVGLS); global myocardial work (GMW) indices, which include global work index (GWI), global constructive work (GCW), global wasted work, and global work efficiency; and the LS of subendocardial, middle, and subepicardial layer myocardium during left ventricular systole were measured: left atrial strain of reservoir phase (LASr), left atrial strain of conduit phase (LAScd), and left atrial strain of contraction phase (LASct). Results One-way ANOVA showed that GLS, GWI, GCW, LASr, and LAScd were closely associated with CTRCD and multivariate logistic regression analysis showed that GLS was the most sensitive predictor for detecting patients at lofty risk of anthracycline-related cardiotoxicity. Both before and after chemotherapy, GLS in the left ventricle showed a pattern of basal segment < middle segment < apical segment and subepicardial < middle < subendocardial layer (p < 0.05), and the degree of decrease also showed a regular pattern of epicardial layer < middle layer < subendocardial layer while the difference was not significant (p > 0.05). After chemotherapy, maximum flow rate in early mitral relaxation/left atrial systolic maximum flow rate (E/A) and left atrial volume index of each group were in the normal range, and the values of LASr, LAScd, and LASct slightly increased in the second cycle and decreased significantly in the fourth cycle after chemotherapy, reaching the lowest level; LASr and LAScd were positively correlated with GLS. Conclusion LVGLS is a more sensitive and earlier indicator to predict CTRCD compared with conventional echocardiography-related parameters and serological markers, and GLS of each myocardial layer showed a certain regularity. Left atrial strain can be used for early monitoring of cardiotoxicity in children with lymphoma after chemotherapy.
Collapse
Affiliation(s)
- Jiaqi Chen
- Department of Ultrasound, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
| | - Chunyue Cheng
- Department of Ultrasound, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Fan
- Department of Ultrasound, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
| | - Xiaochuan Xu
- Department of Ultrasound, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
| | - Jing Chen
- Department of Ultrasound, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
| | - Yang Feng
- Department of Ultrasound, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
| | - Yi Tang
- Department of Ultrasound, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
| | - Chunjiang Yang
- Department of Ultrasound, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
| |
Collapse
|
67
|
Pan J, Xiong W, Zhang A, Zhang H, Lin H, Gao L, Ke J, Huang S, Zhang J, Gu J, Chang ACY, Wang C. The Imbalance of p53-Park7 Signaling Axis Induces Iron Homeostasis Dysfunction in Doxorubicin-Challenged Cardiomyocytes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206007. [PMID: 36967569 DOI: 10.1002/advs.202206007] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 02/23/2023] [Indexed: 05/27/2023]
Abstract
Doxorubicin (DOX)-induced cardiotoxicity (DoIC) is a major side effect for cancer patients. Recently, ferroptosis, triggered by iron overload, is demonstrated to play a role in DoIC. How iron homeostasis is dysregulated in DoIC remains to be elucidated. Here, the authors demonstrate that DOX challenge exhibits reduced contractile function and induction of ferroptosis-related phenotype in cardiomyocytes, evidenced by iron overload, lipid peroxide accumulation, and mitochondrial dysfunction. Compared to Ferric ammonium citrate (FAC) induced secondary iron overload, DOX-challenged cardiomyocytes show a dysfunction of iron homeostasis, with decreased cytoplasmic and mitochondrial iron-sulfur (FeS) cluster-mediated aconitase activity and abnormal expression of iron homeostasis-related proteins. Mechanistically, mass spectrometry analysis identified DOX-treatment induces p53-dependent degradation of Parkinsonism associated deglycase (Park7) which results in iron homeostasis dysregulation. Park7 counteracts iron overload by regulating iron regulatory protein family transcription while blocking mitochondrial iron uptake. Knockout of p53 or overexpression of Park7 in cardiomyocytes remarkably restores the activity of FeS cluster and iron homeostasis, inhibits ferroptosis, and rescues cardiac function in DOX treated animals. These results demonstrate that the iron homeostasis plays a key role in DoIC ferroptosis. Targeting of the newly identified p53-Park7 signaling axis may provide a new approach to prevent DoIC.
Collapse
Affiliation(s)
- Jianan Pan
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University school of Medicine, Shanghai, 200001, P. R. China
| | - Weiyao Xiong
- Department of Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai JiaoTong University school of Medicine, Shanghai, 200135, P. R. China
| | - Alian Zhang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University school of Medicine, Shanghai, 200001, P. R. China
| | - Hui Zhang
- Department of Echocardiography, Zhongshan Hospital, Fudan University, Shanghai, 200030, P. R. China
| | - Hao Lin
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University school of Medicine, Shanghai, 200001, P. R. China
| | - Lin Gao
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University school of Medicine, Shanghai, 200001, P. R. China
| | - Jiahan Ke
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University school of Medicine, Shanghai, 200001, P. R. China
| | - Shuying Huang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University school of Medicine, Shanghai, 200001, P. R. China
| | - Junfeng Zhang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University school of Medicine, Shanghai, 200001, P. R. China
| | - Jun Gu
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University school of Medicine, Shanghai, 200001, P. R. China
| | - Alex Chia Yu Chang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University school of Medicine, Shanghai, 200001, P. R. China
- Department of Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai JiaoTong University school of Medicine, Shanghai, 200135, P. R. China
| | - Changqian Wang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University school of Medicine, Shanghai, 200001, P. R. China
| |
Collapse
|
68
|
Wang Q, Jiang F, Zhao C, Song J, Hu M, Lv Y, Duan Y, Fang W, Ding R, Qiu Y. miR-21-5p prevents doxorubicin-induced cardiomyopathy by downregulating BTG2. Heliyon 2023; 9:e15451. [PMID: 37131441 PMCID: PMC10149273 DOI: 10.1016/j.heliyon.2023.e15451] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/27/2023] [Accepted: 04/10/2023] [Indexed: 05/04/2023] Open
Abstract
Cardiomyocyte apoptosis has been characterized as one of the major mechanisms underlying doxorubicin (DOX)-induced cardiomyopathy. MicroRNA-21-5p (miR-21-5p) was reported to mitigate ischemia-induced cardiomyocyte apoptosis and cardiac injury. However, to our knowledge, the functional role of miR-21-5p in DOX-induced cardiomyopathy is unclear. In this study, we explored the role of miR-21-5p in DOX-induced cardiac injury. The expression level of miR-21-5p was detected by quantitative real-time polymerase chain reaction (qRT-PCR). Dual luciferase reporter assay was used to verify the potential target gene of miR-21-5p. The apoptosis rate of NRCMs was detected by TUNEL staining assay. Western blot analysis was used to detect the protein expression levels of Bax, Bcl-2, Caspase3, cleaved-Caspase3 and BTG2. For animal studies, mice were injected with AAV9-miR-21-5p or AAV9-Empty viruses, and treated with DOX at a dose of 5 mg/kg per week through intraperitoneally administration. After 4 weeks of DOX treatment, mice were subjected to echocardiography to measure the left ventricular ejection fraction (EF) and fractional shortening (FS). Results showed that miR-21-5p was upregulated in both DOX-treated primary cardiomyocytes and mouse heart tissues. Interestingly, enhanced miR-21-5p expression inhibited DOX-induced cardiomyocyte apoptosis and oxidative stress, while decreased miR-21-5p expression promoted cardiomyocyte apoptosis and oxidative stress. Furthermore, cardiac overexpression of miR-21-5p protected against DOX-induced cardiac injury. The mechanistic study indicated that BTG2 was a target gene of miR-21-5p. The anti-apoptotic effect of miR-21-5p could be inhibited by BTG2 overexpression. Conversely, inhibition of BTG2 rescued the pro-apoptotic effect of miR-21-5p inhibitor. Taken together, our study showed that miR-21-5p could prevent DOX-induced cardiomyopathy by downregulating BTG2.
Collapse
Affiliation(s)
- Qingwei Wang
- Department of Cardiology, People's Hospital, Peking University, Beijing, 100044, China
| | - Fei Jiang
- Heart Medicine Research Center, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| | - Chenglin Zhao
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Jiaxin Song
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Meiyu Hu
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Yicheng Lv
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Yi Duan
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Wenqian Fang
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Rongjing Ding
- Cardiac Rehabilitation Center, Department of Rehabilitation Medicine, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Yan Qiu
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
| |
Collapse
|
69
|
Recent progress in nanocarrier-based drug delivery systems for antitumour metastasis. Eur J Med Chem 2023; 252:115259. [PMID: 36934485 DOI: 10.1016/j.ejmech.2023.115259] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 02/24/2023] [Accepted: 03/07/2023] [Indexed: 03/14/2023]
Abstract
Tumour metastasis is one of the major factors leading to poor prognosis as well as lower survival among cancer patients. A number of studies investigating the inhibition of tumour metastasis have been conducted. It is difficult to achieve satisfactory results with surgery alone for distant metastatic tumours, and chemotherapy can boost the healing rate and prognosis of patients. However, the poor therapeutic efficacy of chemotherapy drugs due to their low solubility, lack of tumour targeting, instability in vivo, high toxicity and multidrug resistance hinder their application. Immunotherapy is beneficial to the treatment of metastatic cancers, but it also has disadvantages such as adverse reactions and acquired resistance. Fortunately, delivery of chemotherapeutic drugs with nanocarriers can reduce systemic reactions caused by chemotherapeutic agents and inhibit metastasis. This review discusses the underlying mechanisms of metastasis, therapeutic approaches for antitumour metastasis, the advantages of nanodrug delivery systems and their application in reducing metastasis.
Collapse
|
70
|
Feng J, Wu Y. Endothelial-to-Mesenchymal Transition: Potential Target of Doxorubicin-Induced Cardiotoxicity. Am J Cardiovasc Drugs 2023; 23:231-246. [PMID: 36841924 DOI: 10.1007/s40256-023-00573-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/02/2023] [Indexed: 02/27/2023]
Abstract
The use of chemotherapeutic agents is becoming more frequent as the proportion of new oncology patients increases worldwide, with prolonged survival after treatment. As one of the most popular chemotherapy drugs, doxorubicin plays a substantial role in the treatment of tumors. Unfortunately, the use of doxorubicin is associated with several adverse effects, particularly severe cardiotoxicity that can be life-threatening, which greatly limits its clinical use. For decades, scientists have tried to explore many cardioprotective agents and therapeutic approaches, but their efficacy remains controversial, and some drugs have even brought about significant adverse effects. The concrete molecular mechanism of doxorubicin-induced cardiotoxicity is still to be unraveled, yet endothelial damage is gradually being identified as an important mechanism triggering the development and progression of doxorubicin-induced cardiotoxicity. Endothelial-to-mesenchymal transition (EndMT), a fundamental process regulating morphogenesis in multicellular organisms, is recognized to be associated with endothelial damage repair and acts as an important factor in the progression of cardiovascular diseases, tumors, and rheumatic immune diseases. Mounting evidence suggests that endothelial-mesenchymal transition may play a non-negligible role in doxorubicin-induced cardiotoxicity. In this paper, we reviewed the molecular mechanisms and signaling pathways of EndMT and outlined the molecular mechanisms of doxorubicin-induced cardiotoxicity and the current therapeutic advances. Furthermore, we summarized the basic principles of doxorubicin-induced endothelial-mesenchymal transition that lead to endothelial dysfunction and cardiotoxicity, aiming to provide suggestions or new ideas for the prevention and treatment of doxorubicin-induced endothelial and cardiac injury.
Collapse
Affiliation(s)
- Jie Feng
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Yanqing Wu
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
71
|
Zhang Y, Liu S, Peng J, Cheng S, Zhang Q, Zhang N, Zhou Z, Zhang Y, Zhao Y, Liu T. Biomimetic Nanozymes Suppressed Ferroptosis to Ameliorate Doxorubicin-Induced Cardiotoxicity via Synergetic Effect of Antioxidant Stress and GPX4 Restoration. Nutrients 2023; 15:1090. [PMID: 36904089 PMCID: PMC10005374 DOI: 10.3390/nu15051090] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 02/15/2023] [Accepted: 02/21/2023] [Indexed: 02/24/2023] Open
Abstract
Mitochondria-dependent ferroptosis plays an important role in the pathogenesis of doxorubicin (DOX)-induced cardiotoxicity (DIC), which remains a clinical challenge due to the lack of effective interventions. Cerium oxide (CeO2), a representative nanozyme, has attracted much attention because of its antioxidant properties. This study evaluated CeO2-based nanozymes for the prevention and treatment of DIC in vitro and in vivo by adding nanoparticles (NPs), which were synthesized by biomineralization, to the culture or giving them to the mice, and the ferroptosis-specific inhibitor ferrostatin-1 (Fer-1) was used as control. The prepared NPs exhibited an excellent antioxidant response and glutathione peroxidase 4 (GPX4)-depended bioregulation, with the additional merits of bio-clearance and long retention in the heart. The experiments showed that NP treatment could significantly reverse myocardial structural and electrical remodeling, and reduce myocardial necrosis. These cardioprotective therapeutic effects were associated with their ability to alleviate oxidative stress, mitochondrial lipid peroxidation, and mitochondrial membrane potential damage, with a superior efficiency to the Fer-1. The study also found that the NPs significantly restored the expression of GPX4 and mitochondrial-associated proteins, thereby restoring mitochondria-dependent ferroptosis. Therefore, the study provides some insights into the role of ferroptosis in DIC. It also shows that CeO2-based nanozymes could be a promising prevention and treatment candidate as a novel cardiomyocyte ferroptosis protector to mitigate DIC and improve prognosis and quality of life in cancer patients.
Collapse
Affiliation(s)
- Yunpeng Zhang
- Department of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Shuang Liu
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Jing Peng
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Shifeng Cheng
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Qingling Zhang
- Department of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Nan Zhang
- Department of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Zandong Zhou
- Department of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Yue Zhang
- Department of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Yang Zhao
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Tong Liu
- Department of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| |
Collapse
|
72
|
Berberine Alleviates Doxorubicin-Induced Myocardial Injury and Fibrosis by Eliminating Oxidative Stress and Mitochondrial Damage via Promoting Nrf-2 Pathway Activation. Int J Mol Sci 2023; 24:ijms24043257. [PMID: 36834687 PMCID: PMC9966753 DOI: 10.3390/ijms24043257] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 01/30/2023] [Accepted: 02/03/2023] [Indexed: 02/11/2023] Open
Abstract
Doxorubicin (DOX)-related cardiotoxicity has been recognized as a serious complication of cancer chemotherapy. Effective targeted strategies for myocardial protection in addition to DOX treatment are urgently needed. The purpose of this paper was to determine the therapeutic effect of berberine (Ber) on DOX-triggered cardiomyopathy and explore the underlying mechanism. Our data showed that Ber markedly prevented cardiac diastolic dysfunction and fibrosis, reduced cardiac malondialdehyde (MDA) level and increased antioxidant superoxide dismutase (SOD) activity in DOX-treated rats. Moreover, Ber effectively rescued the DOX-induced production of reactive oxygen species (ROS) and MDA, mitochondrial morphological damage and membrane potential loss in neonatal rat cardiac myocytes and fibroblasts. This effect was mediated by increases in the nuclear accumulation of nuclear erythroid factor 2-related factor 2 (Nrf2) and levels of heme oxygenase-1 (HO-1) and mitochondrial transcription factor A (TFAM). We also found that Ber suppressed the differentiation of cardiac fibroblasts (CFs) into myofibroblasts, as indicated by decreased expression of α-smooth muscle actin (α-SMA), collagen I and collagen III in DOX-treated CFs. Pretreatment with Ber inhibited ROS and MDA production and increased SOD activity and the mitochondrial membrane potential in DOX-challenged CFs. Further investigation indicated that the Nrf2 inhibitor trigonelline reversed the protective effect of Ber on both cardiomyocytes and CFs after DOX stimulation. Taken together, these findings demonstrated that Ber effectively alleviated DOX-induced oxidative stress and mitochondrial damage by activating the Nrf2-mediated pathway, thereby leading to the prevention of myocardial injury and fibrosis. The current study suggests that Ber is a potential therapeutic agent for DOX-induced cardiotoxicity that exerts its effects by activating Nrf2.
Collapse
|
73
|
Abstract
The cardiovascular system requires iron to maintain its high energy demands and metabolic activity. Iron plays a critical role in oxygen transport and storage, mitochondrial function, and enzyme activity. However, excess iron is also cardiotoxic due to its ability to catalyze the formation of reactive oxygen species and promote oxidative damage. While mammalian cells have several redundant iron import mechanisms, they are equipped with a single iron-exporting protein, which makes the cardiovascular system particularly sensitive to iron overload. As a result, iron levels are tightly regulated at many levels to maintain homeostasis. Iron dysregulation ranges from iron deficiency to iron overload and is seen in many types of cardiovascular disease, including heart failure, myocardial infarction, anthracycline-induced cardiotoxicity, and Friedreich's ataxia. Recently, the use of intravenous iron therapy has been advocated in patients with heart failure and certain criteria for iron deficiency. Here, we provide an overview of systemic and cellular iron homeostasis in the context of cardiovascular physiology, iron deficiency, and iron overload in cardiovascular disease, current therapeutic strategies, and future perspectives.
Collapse
Affiliation(s)
- Konrad Teodor Sawicki
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University, Chicago, IL 60611
- Division of Cardiology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Adam De Jesus
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University, Chicago, IL 60611
| | - Hossein Ardehali
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University, Chicago, IL 60611
- Division of Cardiology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| |
Collapse
|
74
|
Wu L, Wang L, Du Y, Zhang Y, Ren J. Mitochondrial quality control mechanisms as therapeutic targets in doxorubicin-induced cardiotoxicity. Trends Pharmacol Sci 2023; 44:34-49. [PMID: 36396497 DOI: 10.1016/j.tips.2022.10.003] [Citation(s) in RCA: 112] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/21/2022] [Accepted: 10/25/2022] [Indexed: 11/16/2022]
Abstract
Doxorubicin (DOX) is a chemotherapeutic drug that is utilized for solid tumors and hematologic malignancies, but its clinical application is hampered by life-threatening cardiotoxicity including cardiac dilation and heart failure. Mitochondrial quality control processes, including mitochondrial proteostasis, mitophagy, and mitochondrial dynamics and biogenesis, serve to maintain mitochondrial homeostasis in the cardiovascular system. Importantly, recent advances have unveiled a major role for defective mitochondrial quality control in the etiology of DOX cardiomyopathy. Moreover, specific interventions targeting these quality control mechanisms to preserve mitochondrial function have emerged as potential therapeutic strategies to attenuate DOX cardiotoxicity. However, clinical translation is challenging because of obscure mechanisms of action and potential adverse effects. The purpose of this review is to provide new insights regarding the role of mitochondrial quality control in the pathogenesis of DOX cardiotoxicity, and to explore promising therapeutic approaches targeting these mechanisms to aid clinical management.
Collapse
Affiliation(s)
- Lin Wu
- Department of Cardiology and Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Litao Wang
- Department of Cardiology and Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yuxin Du
- Department of Cardiology and Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yingmei Zhang
- Department of Cardiology and Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jun Ren
- Department of Cardiology and Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| |
Collapse
|
75
|
Zhou P, Gao G, Zhao CC, Li JY, Peng JF, Wang SS, Song R, Shi H, Wang L. In vivo and in vitro protective effects of shengmai injection against doxorubicin-induced cardiotoxicity. PHARMACEUTICAL BIOLOGY 2022; 60:638-651. [PMID: 35298357 PMCID: PMC8933025 DOI: 10.1080/13880209.2022.2046801] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 02/07/2022] [Accepted: 02/21/2022] [Indexed: 06/14/2023]
Abstract
CONTEXT Shengmai injection (SMI) has been used to treat heart failure. OBJECTIVE This study determines the molecular mechanisms of SMI against cardiotoxicity caused by doxorubicin (DOX). MATERIALS AND METHODS In vivo, DOX (15 mg/kg) was intraperitoneally injected in model, Dex (dexrazoxane), SMI-L (2.7 mL/kg), SMI-M (5.4 mL/kg), and SMI-H (10.8 mL/kg) for 7 consecutive days. Hematoxylin-eosin (HE) and Masson staining were used to evaluate histological changes, and cardiomyocyte apoptosis was identified using TdT-mediated dUTP nick-end labelling (TUNEL). Enzymatic indexes were determined. mRNA and protein expressions were analysed through RT-qPCR and Western blotting. In vitro, H9c2 cells were divided into control group, model group (2 mL 1 μM DOX), SMI group, ML385 group, and SMI + ML385 group, the intervention lasted for 24 h. mRNA and protein expressions were analysed. RESULTS SMI markedly improved cardiac pathology, decreased cardiomyocyte apoptosis, increased creatine kinase (CK), lactate dehydrogenase (LDH), malondialdehyde (MDA), decreased superoxide dismutase (SOD). Compared with the model group, the protein expression of nuclear factor erythroid2-related factor 2 (Nrf2) (SMI-L: 2.42-fold, SMI-M: 2.67-fold, SMI-H: 3.07-fold) and haem oxygenase-1(HO-1) (SMI-L: 1.64-fold, SMI-M: 2.01-fold, SMI-H: 2.19-fold) was increased and the protein expression of kelch-like ECH-associated protein 1 (Keap1) (SMI-L: 0.90-fold, SMI-M: 0.77-fold, SMI-H: 0.66-fold) was decreased in SMI groups and Dex group in vivo. Additionally, SMI dramatically inhibited apoptosis, decreased CK, LDH and MDA levels, and enhanced SOD activity. Our results demonstrated that SMI reduced DOX-induced cardiotoxicity via activation of the Nrf2/Keap1 signalling pathway. CONCLUSIONS This study revealed a new mechanism by which SMI alleviates DOX-induced 45 cardiomyopathy by modulating the Nrf2/Keap1 signal pathway.
Collapse
Affiliation(s)
- Peng Zhou
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
- Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Ge Gao
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Chun-chun Zhao
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Jing-ya Li
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Jian-fei Peng
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Shu-shu Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Rui Song
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Hui Shi
- Nursing School, Anhui University of Chinese Medicine, Hefei, China
| | - Liang Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
- Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| |
Collapse
|
76
|
Affiliation(s)
- Carlo G Tocchetti
- Cardio-Oncology Unit, Department of Translational Medical Sciences (DISMET), Center for Basic and Clinical Immunology Research (CISI), Interdepartmental Center of Clinical and Translational Sciences (CIRCET), Interdepartmental Hypertension Research Center (CIRIAPA), Federico II University, Naples, Italy
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126 Torino, Italy
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126 Torino, Italy
| |
Collapse
|
77
|
Effect of aerobic exercise on cardiotoxic outcomes in women with breast cancer undergoing anthracycline or trastuzumab treatment: a systematic review and meta-analysis. Support Care Cancer 2022; 30:10323-10334. [DOI: 10.1007/s00520-022-07368-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 09/18/2022] [Indexed: 11/05/2022]
|
78
|
Ling G, Wang X, Tan N, Cao J, Li W, Zhang Y, Jiang J, Sun Q, Jiang Y, Wang W, Wang Y. Mechanisms and Drug Intervention for Doxorubicin-Induced Cardiotoxicity Based on Mitochondrial Bioenergetics. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7176282. [PMID: 36275901 PMCID: PMC9586735 DOI: 10.1155/2022/7176282] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 06/17/2022] [Accepted: 09/10/2022] [Indexed: 11/18/2022]
Abstract
Doxorubicin (DOX) is an anthracycline chemotherapy drug, which is indispensable in antitumor therapy. However, its subsequent induction of cardiovascular disease (CVD) has become the primary cause of mortality in cancer survivors. Accumulating evidence has demonstrated that cardiac mitochondrial bioenergetics changes have become a significant marker for doxorubicin-induced cardiotoxicity (DIC). Here, we mainly summarize the related mechanisms of DOX-induced cardiac mitochondrial bioenergetics disorders reported in recent years, including mitochondrial substrate metabolism, the mitochondrial respiratory chain, myocardial ATP storage and utilization, and other mechanisms affecting mitochondrial bioenergetics. In addition, intervention for DOX-induced cardiac mitochondrial bioenergetics disorders using chemical drugs and traditional herbal medicine is also summarized, which will provide a comprehensive process to study and develop more appropriate therapeutic strategies for DIC.
Collapse
Affiliation(s)
- Guanjing Ling
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xiaoping Wang
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Nannan Tan
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jing Cao
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Weili Li
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yawen Zhang
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jinchi Jiang
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Qianbin Sun
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yanyan Jiang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Wei Wang
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
- Beijing Key Laboratory of TCM Syndrome and Formula, Beijing 100029, China
- Key Laboratory of Beijing University of Chinese Medicine, Ministry of Education, Beijing 100029, China
- Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Yong Wang
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
- Beijing Key Laboratory of TCM Syndrome and Formula, Beijing 100029, China
- Key Laboratory of Beijing University of Chinese Medicine, Ministry of Education, Beijing 100029, China
| |
Collapse
|
79
|
Li W, Li S, Cao Z, Sun Y, Qiu W, Jia M, Su M. Exploration of the amino acid metabolic signature in anthracycline-induced cardiotoxicity using an optimized targeted metabolomics approach based on UPLC-MS/MS. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2022; 395:1209-1224. [PMID: 35879430 DOI: 10.1007/s00210-022-02271-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 07/08/2022] [Indexed: 10/16/2022]
Abstract
Although anthracyclines improve the long-term survival rate of patients with cancer, severe and irreversible myocardial damage limits their clinical application. Amino acid (AA) metabolism in cardiomyocytes can be altered under pathological conditions. Therefore, exploring the AA metabolic signature in anthracycline-induced cardiotoxicity (AIC) is important for identifying novel mechanisms. We established mouse and cellular models of Adriamycin (ADR)-induced cardiac injury. We observed a decreased expression of troponins I (cTnI) after ADR treatment and ADR accelerated the degradation of cTnI, implying that AA metabolism could be altered in AIC. Using a targeted AA metabolomics approach based on ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS), the AA metabolic signatures in the sera of AIC mice and supernatant samples of ADR-treated H9c2 cardiomyocytes were analyzed. The levels of 14 AA metabolites were altered in ADR-treated mice (p < 0.05). Via bioinformatics analysis, we identified nine differential AA metabolites in mice and five differential AA metabolites in ADR-treated H9c2 cardiomyocytes. Three AAs with increased levels (L-glutamate, L-serine, and L-tyrosine) overlapped in the two models, suggesting a possible mechanism of AA metabolic impairment during AIC. The metabolic pathways perturbed by AIC involved aminoacyl-tRNA biosynthesis and alanine, aspartate, and glutamate metabolism. Our data suggests that ADR perturbed AA metabolism in AIC models. Moreover, the targeted AA metabolomics approach based on UPLC-MS/MS can be a unique platform to provide new clues for the prevention and treatment of AIC.
Collapse
Affiliation(s)
- Wendi Li
- Department of Clinical Laboratory, Peking University People's Hospital, No. 11 Xizhimen South Street, Beijing, 100044, People's Republic of China
| | - Shanshan Li
- Department of Clinical Laboratory, Peking University People's Hospital, No. 11 Xizhimen South Street, Beijing, 100044, People's Republic of China
| | - Zhenju Cao
- Department of Clinical Laboratory, Peking University People's Hospital, No. 11 Xizhimen South Street, Beijing, 100044, People's Republic of China
| | - Yi Sun
- Department of Clinical Laboratory, Peking University People's Hospital, No. 11 Xizhimen South Street, Beijing, 100044, People's Republic of China
| | - Wei Qiu
- Department of Urology, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, People's Republic of China.
| | - Mei Jia
- Department of Clinical Laboratory, Peking University People's Hospital, No. 11 Xizhimen South Street, Beijing, 100044, People's Republic of China.
| | - Ming Su
- Department of Clinical Laboratory, Peking University People's Hospital, No. 11 Xizhimen South Street, Beijing, 100044, People's Republic of China.
| |
Collapse
|
80
|
Li MJ, Sun WS, Yuan Y, Zhang YK, Lu Q, Gao YZ, Ye T, Xing DM. Breviscapine remodels myocardial glucose and lipid metabolism by regulating serotonin to alleviate doxorubicin-induced cardiotoxicity. Front Pharmacol 2022; 13:930835. [PMID: 36238546 PMCID: PMC9551275 DOI: 10.3389/fphar.2022.930835] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 08/22/2022] [Indexed: 11/22/2022] Open
Abstract
Aims: The broad-spectrum anticancer drug doxorubicin (Dox) is associated with a high incidence of cardiotoxicity, which severely affects the clinical application of the drug and patients’ quality of life. Here, we assess how Dox modulates myocardial energy and contractile function and this could aid the development of relevant protective drugs. Methods: Mice were subjected to doxorubicin and breviscapine treatment. Cardiac function was analyzed by echocardiography, and Dox-mediated signaling was assessed in isolated cardiomyocytes. The dual cardio-protective and anti-tumor actions of breviscapine were assessed in mouse breast tumor models. Results: We found that Dox disrupts myocardial energy metabolism by decreasing glucose uptake and increasing fatty acid oxidation, leading to a decrease in ATP production rate, an increase in oxygen consumption rate and oxidative stress, and further energy deficits to enhance myocardial fatty acid uptake and drive DIC development. Interestingly, breviscapine increases the efficiency of ATP production and restores myocardial energy homeostasis by modulating the serotonin-glucose-myocardial PI3K/AKT loop, increasing glucose utilization by the heart and reducing lipid oxidation. It enhances mitochondrial autophagy via the PINK1/Parkin pathway, eliminates damaged mitochondrial accumulation caused by Dox, reduces the degree of cardiac fibrosis and inflammation, and restores cardiac micro-environmental homeostasis. Importantly, its low inflammation levels reduce myeloid immunosuppressive cell infiltration, and this effect is synergistic with the anti-tumor effect of Dox. Conclusion: Our findings suggest that disruption of the cardiac metabolic network by Dox is an important driver of its cardiotoxicity and that serotonin is an important regulator of myocardial glucose and lipid metabolism. Myocardial energy homeostasis and timely clearance of damaged mitochondria synergistically contribute to the prevention of anthracycline-induced cardiotoxicity and improve the efficiency of tumor treatment.
Collapse
Affiliation(s)
- Meng-Jiao Li
- Cancer Institute of the Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Wen-She Sun
- Cancer Institute of the Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
| | - Yang Yuan
- Cancer Institute of the Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
| | - Yu-Kun Zhang
- Cancer Institute of the Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Qi Lu
- Cancer Institute of the Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Yuan-Zhen Gao
- Cancer Institute of the Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Ting Ye
- Cancer Institute of the Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Dong-Ming Xing
- Cancer Institute of the Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
- School of Life Sciences, Tsinghua University, Beijing, China
- *Correspondence: Dong-Ming Xing,
| |
Collapse
|
81
|
Moro N, Dokshokova L, Perumal Vanaja I, Prando V, Cnudde SJA, Di Bona A, Bariani R, Schirone L, Bauce B, Angelini A, Sciarretta S, Ghigo A, Mongillo M, Zaglia T. Neurotoxic Effect of Doxorubicin Treatment on Cardiac Sympathetic Neurons. Int J Mol Sci 2022; 23:ijms231911098. [PMID: 36232393 PMCID: PMC9569551 DOI: 10.3390/ijms231911098] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/14/2022] [Accepted: 09/17/2022] [Indexed: 11/26/2022] Open
Abstract
Doxorubicin (DOXO) remains amongst the most commonly used anti-cancer agents for the treatment of solid tumors, lymphomas, and leukemias. However, its clinical use is hampered by cardiotoxicity, characterized by heart failure and arrhythmias, which may require chemotherapy interruption, with devastating consequences on patient survival and quality of life. Although the adverse cardiac effects of DOXO are consolidated, the underlying mechanisms are still incompletely understood. It was previously shown that DOXO leads to proteotoxic cardiomyocyte (CM) death and myocardial fibrosis, both mechanisms leading to mechanical and electrical dysfunction. While several works focused on CMs as the culprits of DOXO-induced arrhythmias and heart failure, recent studies suggest that DOXO may also affect cardiac sympathetic neurons (cSNs), which would thus represent additional cells targeted in DOXO-cardiotoxicity. Confocal immunofluorescence and morphometric analyses revealed alterations in SN innervation density and topology in hearts from DOXO-treated mice, which was consistent with the reduced cardiotropic effect of adrenergic neurons in vivo. Ex vivo analyses suggested that DOXO-induced denervation may be linked to reduced neurotrophic input, which we have shown to rely on nerve growth factor, released from innervated CMs. Notably, similar alterations were observed in explanted hearts from DOXO-treated patients. Our data demonstrate that chemotherapy cardiotoxicity includes alterations in cardiac innervation, unveiling a previously unrecognized effect of DOXO on cardiac autonomic regulation, which is involved in both cardiac physiology and pathology, including heart failure and arrhythmias.
Collapse
Affiliation(s)
- Nicola Moro
- Department of Biomedical Sciences, University of Padova, via Ugo Bassi 58/B, 35131 Padova, Italy
| | - Lolita Dokshokova
- Department of Biomedical Sciences, University of Padova, via Ugo Bassi 58/B, 35131 Padova, Italy
| | - Induja Perumal Vanaja
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, via Giustiniani 2, 35128 Padova, Italy
| | - Valentina Prando
- Department of Biomedical Sciences, University of Padova, via Ugo Bassi 58/B, 35131 Padova, Italy
| | - Sophie Julie A Cnudde
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | - Anna Di Bona
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, via Giustiniani 2, 35128 Padova, Italy
| | - Riccardo Bariani
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, via Giustiniani 2, 35128 Padova, Italy
| | - Leonardo Schirone
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza, University of Rome, 04100 Latina, Italy
| | - Barbara Bauce
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, via Giustiniani 2, 35128 Padova, Italy
| | - Annalisa Angelini
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, via Giustiniani 2, 35128 Padova, Italy
| | - Sebastiano Sciarretta
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza, University of Rome, 04100 Latina, Italy
| | - Alessandra Ghigo
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | - Marco Mongillo
- Department of Biomedical Sciences, University of Padova, via Ugo Bassi 58/B, 35131 Padova, Italy
- Correspondence: (M.M.); (T.Z.); Tel.: +39-0497923229 (M.M.); +39-0497923294 (T.Z.); Fax: +39-0497923250 (M.M.); +39-0497923250 (T.Z.)
| | - Tania Zaglia
- Department of Biomedical Sciences, University of Padova, via Ugo Bassi 58/B, 35131 Padova, Italy
- Correspondence: (M.M.); (T.Z.); Tel.: +39-0497923229 (M.M.); +39-0497923294 (T.Z.); Fax: +39-0497923250 (M.M.); +39-0497923250 (T.Z.)
| |
Collapse
|
82
|
Kong L, Zhang Y, Ning J, Xu C, Wang Z, Yang J, Yang L. CaMKII
orchestrates endoplasmic reticulum stress and apoptosis in doxorubicin‐induced cardiotoxicity by regulating the
IRE1α
/
XBP1s
pathway. J Cell Mol Med 2022; 26:5303-5314. [PMID: 36111515 PMCID: PMC9575131 DOI: 10.1111/jcmm.17560] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 08/16/2022] [Accepted: 09/05/2022] [Indexed: 11/29/2022] Open
Affiliation(s)
- Lingheng Kong
- Department of Anaesthesiology Xi'an Children's Hospital Xi'an China
- Institute of Basic Medical Science Xi'an Medical University Xi'an China
| | - Yimeng Zhang
- Institute of Basic Medical Science Xi'an Medical University Xi'an China
| | - Jiayi Ning
- Institute of Basic Medical Science Xi'an Medical University Xi'an China
| | - Chennian Xu
- Department of Cardiovascular Surgery, Xijing Hospital Air Force Medical University Xi'an China
- Department of Cardiovascular Surgery General Hospital of Northern Theatre Command Shenyang China
| | - Zhenyi Wang
- Department of Anaesthesiology Xi'an Children's Hospital Xi'an China
| | - Jian Yang
- Department of Cardiovascular Surgery, Xijing Hospital Air Force Medical University Xi'an China
| | - Lifang Yang
- Department of Anaesthesiology Xi'an Children's Hospital Xi'an China
| |
Collapse
|
83
|
Menilli L, Milani C, Reddi E, Moret F. Overview of Nanoparticle-Based Approaches for the Combination of Photodynamic Therapy (PDT) and Chemotherapy at the Preclinical Stage. Cancers (Basel) 2022; 14:cancers14184462. [PMID: 36139623 PMCID: PMC9496990 DOI: 10.3390/cancers14184462] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 09/06/2022] [Accepted: 09/09/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary The present review represents the outstanding and promising recent literature reports (2017–2022) on nanoparticle-based formulations developed for anticancer therapy with photodynamic therapy (PDT), photosensitizers, and chemotherapeutics. Besides brief descriptions of chemotherapeutics’ classification and of PDT mechanisms and limitations, several examples of nanosystems endowed with different responsiveness (e.g., acidic pH and reactive oxygen species) and peculiarity (e.g., tumor oxygenation capacity, active tumor targeting, and biomimetic features) are described, and for each drug combination, in vitro and in vivo results on preclinical cancer models are reported. Abstract The widespread diffusion of photodynamic therapy (PDT) as a clinical treatment for solid tumors is mainly limited by the patient’s adverse reaction (skin photosensivity), insufficient light penetration in deeply seated neoplastic lesions, unfavorable photosensitizers (PSs) biodistribution, and photokilling efficiency due to PS aggregation in biological environments. Despite this, recent preclinical studies reported on successful combinatorial regimes of PSs with chemotherapeutics obtained through the drugs encapsulation in multifunctional nanometric delivery systems. The aim of the present review deals with the punctual description of several nanosystems designed not only with the objective of co-transporting a PS and a chemodrug for combination therapy, but also with the goal of improving the therapeutic efficacy by facing the main critical issues of both therapies (side effects, scarce tumor oxygenation and light penetration, premature drug clearance, unspecific biodistribution, etc.). Therefore, particular attention is paid to the description of bio-responsive drugs and nanoparticles (NPs), targeted nanosystems, biomimetic approaches, and upconverting NPs, including analyzing the therapeutic efficacy of the proposed photo-chemotherapeutic regimens in in vitro and in vivo cancer models.
Collapse
Affiliation(s)
- Luca Menilli
- Department of Biology, University of Padova, 35100 Padova, Italy
| | - Celeste Milani
- Department of Biology, University of Padova, 35100 Padova, Italy
- Institute of Organic Synthesis and Photoreactivity, ISOF-CNR, 40129 Bologna, Italy
| | - Elena Reddi
- Department of Biology, University of Padova, 35100 Padova, Italy
- Correspondence: (E.R.); (F.M.)
| | - Francesca Moret
- Department of Biology, University of Padova, 35100 Padova, Italy
- Correspondence: (E.R.); (F.M.)
| |
Collapse
|
84
|
Qu PR, Jiang ZL, Song PP, Liu LC, Xiang M, Wang J. Saponins and their derivatives: Potential candidates to alleviate anthracycline-induced cardiotoxicity and multidrug resistance. Pharmacol Res 2022; 182:106352. [PMID: 35835369 DOI: 10.1016/j.phrs.2022.106352] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 07/05/2022] [Accepted: 07/08/2022] [Indexed: 10/17/2022]
Abstract
Anthracyclines (ANTs) continue to play an irreplaceable role in oncology treatment. However, the clinical application of ANTs has been limited. In the first place, ANTs can cause dose-dependent cardiotoxicity such as arrhythmia, cardiomyopathy, and congestive heart failure. In the second place, the development of multidrug resistance (MDR) leads to their chemotherapeutic failure. Oncology cardiologists are urgently searching for agents that can both protect the heart and reverse MDR without compromising the antitumor effects of ANTs. Based on in vivo and in vitro data, we found that natural compounds, including saponins, may be active agents for other both natural and chemical compounds in the inhibition of anthracycline-induced cardiotoxicity (AIC) and the reversal of MDR. In this review, we summarize the work of previous researchers, describe the mechanisms of AIC and MDR, and focus on revealing the pharmacological effects and potential molecular targets of saponins and their derivatives in the inhibition of AIC and the reversal of MDR, aiming to encourage future research and clinical trials.
Collapse
Affiliation(s)
- Pei-Rong Qu
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| | - Zhi-Lin Jiang
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| | - Ping-Ping Song
- Institute of Chinese Materia Medica, China Academy of Chinese Medicine Sciences, Beijing 100013, China
| | - Lan-Chun Liu
- Beijing University of traditional Chinese Medicine, Beijing 100029, China
| | - Mi Xiang
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| | - Jie Wang
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| |
Collapse
|
85
|
Zhang K, Du Z, Yuan T, Huang J, Zhao X, Mi S. Long-term cultured microvascular networks on chip for tumor vascularization research and drug testing. BIOMICROFLUIDICS 2022; 16:044101. [PMID: 35845724 PMCID: PMC9282889 DOI: 10.1063/5.0090027] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 06/13/2022] [Indexed: 06/15/2023]
Abstract
The vascular structure of the tumor microenvironment (TME) plays an essential role in the process of metastasis. In vitro microvascular structures that can be maintained for a long time will greatly promote metastasis research. In this study, we constructed a mimicking breast cancer invasion model based on a microfluidic chip platform, and the maintenance time of the self-assembled microvascular networks significantly improved by culturing with fibroblasts (up to 13 days). Using this model, we quantified the invasion ability of breast cancer cells and angiogenesis sprouts caused by cancer cells, and the intravasation behavior of cancer cells was also observed in sprouts. We found that cancer cells could significantly cause angiogenesis by promoting sprouting behaviors of the self-assembled human umbilical vein endothelial cells, which, in turn, promoted the invasion behavior of cancer cells. The drug test results showed that the drug resistance of the widely used anti-cancer drugs 5-Fluorouracil (5-FU) and Doxorubicin (DOX) in the 3D model was higher than that in the 2D model. Meanwhile, we also proved that 5-FU and DOX had the effect of destroying tumor blood vessels. The anti-angiogenic drug Apatinib (VEGFR inhibitor) enhanced the drug effect of DOX on MDA-MB-231 cells, further proving the promoting effect of angiogenesis on the invasion ability of cancer cells. These results indicate that our model is of great value in reconstructing TME and drug testing in vitro.
Collapse
Affiliation(s)
- Ke Zhang
- Open FIESTA Center, International Graduate School at Shenzhen, Tsinghua University, Shenzhen, China
| | - Zhichang Du
- College of Mechanical and Energy Engineering, Jimei University, Xiamen, China
| | - Tianying Yuan
- Precision Medicine and Healthcare Research Center, Tsinghua-Berkeley Shenzhen Institute, Shenzhen, China
| | - Jiajun Huang
- Bio-manufacturing Engineering Laboratory, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China
| | - Xiaoyu Zhao
- Open FIESTA Center, International Graduate School at Shenzhen, Tsinghua University, Shenzhen, China
| | - Shengli Mi
- Author to whom correspondence should be addressed:
| |
Collapse
|
86
|
Maraldo MV, Levis M, Andreis A, Armenian S, Bates J, Brady J, Ghigo A, Lyon AR, Manisty C, Ricardi U, Aznar MC, Filippi AR. An integrated approach to cardioprotection in lymphomas. Lancet Haematol 2022; 9:e445-e454. [PMID: 35512725 DOI: 10.1016/s2352-3026(22)00082-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/03/2022] [Accepted: 03/10/2022] [Indexed: 10/18/2022]
Abstract
In potentially curable cancers, long-term survival depends not only on the successful treatment of the malignancy but also on the risks associated with treatment-related toxicity, especially cardiotoxicity. Malignant lymphomas affect patients at any age, with acute and late toxicity risks that could have a severe effect on morbidity, mortality, and quality of life. Although our understanding of chemotherapy-associated and radiotherapy-associated cardiovascular disease has advanced considerably, new drugs with potential cardiotoxicity have been introduced for the treatment of lymphomas. In this Review, we summarise the mechanisms of treatment-related cardiac injury, available clinical data, and protocols for optimising cardioprotection in lymphomas. We discuss ongoing research strategies to advance our knowledge of the molecular basis of drug-induced and radiation-induced toxicity. Additionally, we emphasise the potential for personalised follow-up and early detection, including the role of biomarkers and novel diagnostic tests, highlighting the role of the cardio-oncology team.
Collapse
Affiliation(s)
- Maja V Maraldo
- Department of Clinical Oncology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Mario Levis
- Department of Clinical Oncology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Alessandro Andreis
- Division of Cardiology, Città della Salute e della Scienza di Torino Hospital, University of Turin, Turin, Italy
| | - Saro Armenian
- Department of Population Sciences, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - James Bates
- Department of Radiation Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Jessica Brady
- Guy's Cancer Centre, Guy's & St Thomas' NHS Foundation Trust, London, UK
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Alexander R Lyon
- Imperial College London and Cardio-oncology Service, Royal Brompton Hospital, London, UK
| | - Charlotte Manisty
- Department of Cardio-oncology, Barts Heart Centre and University College London, London, UK
| | | | - Marianne C Aznar
- Division of Cancer Sciences, Faculty of Biology, Medicine, and Health, University of Manchester and Department of Radiotherapy-Related Research, The Christie NHS, Manchester, UK.
| | - Andrea Riccardo Filippi
- Radiation Oncology, Fondazione IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy
| |
Collapse
|
87
|
Promoting mitochondrial fusion in doxorubicin-induced cardiotoxicity: a novel therapeutic target for cardioprotection. Clin Sci (Lond) 2022; 136:841-860. [PMID: 35543245 DOI: 10.1042/cs20220074] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 05/06/2022] [Accepted: 05/10/2022] [Indexed: 12/06/2022]
Abstract
Changes in mitochondrial dynamics have been recognized as being one of the mechanisms related to cardiotoxicity following a high cumulative dose of doxorubicin (DOX). A mitochondrial division inhibitor (Mdivi-1) and fusion promoter (M1) have been shown to be cardioprotective in a variety of cardiovascular settings, however their anti-cardiotoxic efficacy against DOX therapy remains unclear. We therefore investigated whether treatment with Mdivi-1 and M1 protect the heart against DOX-induced cardiotoxicity via mitochondria-targeted pathways. Male Wistar rats (n=40) received DOX (3 mg/kg, 6 doses, n=32) or 3% DMSO in the normal saline solution (NSS) (n=8) as a control. DOX-injected rats were given one of four treatments beginning with the first DOX injection via intraperitoneal injection: 1) 3% DMSO in NSS (n=8), 2) Mdivi-1 (1.2 mg/kg/day, n=8), 3) M1 (2 mg/kg/day, n=8), and 4) Mdivi-1+M1 (n=8) for 30 days. Cardiac function, mitochondrial function, oxidative stress, myocardial injury, and protein expression associated with inflammation, autophagy, mitophagy, apoptosis and mitochondrial dynamics were determined. DOX caused a significant deterioration in mitochondrial function and dynamic regulation, and an increase in markers of oxidative stress, inflammation, myocardial injury, apoptosis, autophagy, and mitophagy, resulting in impaired cardiac function. Co-treatment of DOX with Mdivi-1, M1, or a combination of the two mitigated these detrimental effects of DOX. These findings imply that either inhibiting fission or promoting fusion of mitochondria protects the heart from DOX-induced myocardial damage. Modulation of mitochondrial dynamics could be a novel therapeutic target in alleviating DOX-induced cytotoxic effects without compromising its anti-cancer efficacy.
Collapse
|
88
|
Li M, Li H, Liu H, Lai X, Xing W. Efficacy of Chinese Medicine Injection for Cardiotoxic Injury of Anthracycline Chemotherapy Drugs: A Network Meta-Analysis of Randomized Controlled Trials. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:5800575. [PMID: 35399632 PMCID: PMC8991398 DOI: 10.1155/2022/5800575] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/05/2022] [Indexed: 11/19/2022]
Abstract
Background Chinese medicine injections (CMIs) are widely used in the prevention and treatment of cardiotoxicity caused by anthracycline chemotherapeutic drugs. However, it is uncertain that CMIs are more effective in combating the cardiotoxicity of anthracyclines. The aim of this Network Meta-analysis (NMA) was to compare the treatment effects of various CMIs in order to determine the best CMI for the prevention and treatment of cardiac damage from anthracyclines. Methods The Chinese Journal Full Text Database (CNKI), Wanfang Database, Chinese Science and Technology Journal Full Text Database (VIP), Chinese Biomedical Literature Database (CBM), PubMed, Web of Science, and Cochrane Library databases were searched to screen randomized controlled trials (RCTs) of CMIs against cardiotoxicity of anthracycline-based chemotherapeutic drugs. The search time frame was all from the establishment of the database to October 1, 2021. After independent screening of the literature, extraction of information and evaluation of the risk of bias of the included studies by two evaluators, mesh meta-analysis was performed using RevMan 5.3, Stata 15.1, and ADDIS 1.16.8 software. Results A total of 33 studies including 2783 patients, including 1410 cases in the experimental group and 1373 cases in the control group were included, and six CMIs were extracted, namely, Shenfu injection, Shenmai injection, Shenqi Fuzheng injection, Shengmai injection, Xinmailong injection, and Haungqi injection. The results of the reticulated meta-analysis showed that in terms of ST-T segment (ECG change) change rate, Haungqi injection, Shenfu injection, and Xinmailong injection were superior. In terms of lowering CK-MB, Huangqi Injection and Shenqi Fuzheng injection were superior. In terms of improving Left ventricular ejection fraction (LVEF), Shenfu injection, Huangqi Injection, and Shengmai injection were more effective than other injections. In terms of improving LVEDD, Shengmai injection, Huangqi Injection, and Xinmailong injection have advantages. Conclusion The six CMIs included in this study are effective against cardiotoxicity caused by anthracycline-based chemotherapeutic agents. Huangqi Injection and Shenfu injection are both superior in improving various outcome indicators. There is still a need for larger, high-quality randomized controlled trials (RCTs) to compare the various CMIs in a more refined way.
Collapse
Affiliation(s)
- Mingxuan Li
- Beijing University of Chinese Medicine, Beijing, China
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Hongdian Li
- Beijing University of Chinese Medicine, Beijing, China
| | - Hongxu Liu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Xiaolei Lai
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Wenlong Xing
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| |
Collapse
|
89
|
Kwan JM, Oikonomou EK, Henry ML, Sinusas AJ. Multimodality Advanced Cardiovascular and Molecular Imaging for Early Detection and Monitoring of Cancer Therapy-Associated Cardiotoxicity and the Role of Artificial Intelligence and Big Data. Front Cardiovasc Med 2022; 9:829553. [PMID: 35369354 PMCID: PMC8964995 DOI: 10.3389/fcvm.2022.829553] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 01/12/2022] [Indexed: 12/12/2022] Open
Abstract
Cancer mortality has improved due to earlier detection via screening, as well as due to novel cancer therapies such as tyrosine kinase inhibitors and immune checkpoint inhibitions. However, similarly to older cancer therapies such as anthracyclines, these therapies have also been documented to cause cardiotoxic events including cardiomyopathy, myocardial infarction, myocarditis, arrhythmia, hypertension, and thrombosis. Imaging modalities such as echocardiography and magnetic resonance imaging (MRI) are critical in monitoring and evaluating for cardiotoxicity from these treatments, as well as in providing information for the assessment of function and wall motion abnormalities. MRI also allows for additional tissue characterization using T1, T2, extracellular volume (ECV), and delayed gadolinium enhancement (DGE) assessment. Furthermore, emerging technologies may be able to assist with these efforts. Nuclear imaging using targeted radiotracers, some of which are already clinically used, may have more specificity and help provide information on the mechanisms of cardiotoxicity, including in anthracycline mediated cardiomyopathy and checkpoint inhibitor myocarditis. Hyperpolarized MRI may be used to evaluate the effects of oncologic therapy on cardiac metabolism. Lastly, artificial intelligence and big data of imaging modalities may help predict and detect early signs of cardiotoxicity and response to cardioprotective medications as well as provide insights on the added value of molecular imaging and correlations with cardiovascular outcomes. In this review, the current imaging modalities used to assess for cardiotoxicity from cancer treatments are discussed, in addition to ongoing research on targeted molecular radiotracers, hyperpolarized MRI, as well as the role of artificial intelligence (AI) and big data in imaging that would help improve the detection and prognostication of cancer-treatment cardiotoxicity.
Collapse
Affiliation(s)
- Jennifer M. Kwan
- Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Evangelos K. Oikonomou
- Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Mariana L. Henry
- Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - Albert J. Sinusas
- Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, United States
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, United States
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
| |
Collapse
|
90
|
Alov P, Al Sharif M, Aluani D, Chegaev K, Dinic J, Divac Rankov A, Fernandes MX, Fusi F, García-Sosa AT, Juvonen R, Kondeva-Burdina M, Padrón JM, Pajeva I, Pencheva T, Puerta A, Raunio H, Riganti C, Tsakovska I, Tzankova V, Yordanov Y, Saponara S. A Comprehensive Evaluation of Sdox, a Promising H2S-Releasing Doxorubicin for the Treatment of Chemoresistant Tumors. Front Pharmacol 2022; 13:831791. [PMID: 35321325 PMCID: PMC8936434 DOI: 10.3389/fphar.2022.831791] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/25/2022] [Indexed: 12/11/2022] Open
Abstract
Sdox is a hydrogen sulfide (H2S)-releasing doxorubicin effective in P-glycoprotein-overexpressing/doxorubicin-resistant tumor models and not cytotoxic, as the parental drug, in H9c2 cardiomyocytes. The aim of this study was the assessment of Sdox drug-like features and its absorption, distribution, metabolism, and excretion (ADME)/toxicity properties, by a multi- and transdisciplinary in silico, in vitro, and in vivo approach. Doxorubicin was used as the reference compound. The in silico profiling suggested that Sdox possesses higher lipophilicity and lower solubility compared to doxorubicin, and the off-targets prediction revealed relevant differences between Dox and Sdox towards several cancer targets, suggesting different toxicological profiles. In vitro data showed that Sdox is a substrate with lower affinity for P-glycoprotein, less hepatotoxic, and causes less oxidative damage than doxorubicin. Both anthracyclines inhibited CYP3A4, but not hERG currents. Unlike doxorubicin, the percentage of zebrafish live embryos at 72 hpf was not affected by Sdox treatment. In conclusion, these findings demonstrate that Sdox displays a more favorable drug-like ADME/toxicity profile than doxorubicin, different selectivity towards cancer targets, along with a greater preclinical efficacy in resistant tumors. Therefore, Sdox represents a prototype of innovative anthracyclines, worthy of further investigations in clinical settings.
Collapse
Affiliation(s)
- Petko Alov
- Department of QSAR and Molecular Modelling, Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Merilin Al Sharif
- Department of QSAR and Molecular Modelling, Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Denitsa Aluani
- Department of Pharmacology, Pharmacotherapy and Toxicology, Faculty of Pharmacy, Medical University of Sofia, Sofia, Bulgaria
| | - Konstantin Chegaev
- Department of Drug Science and Technology, University of Torino, Torino, Italy
| | - Jelena Dinic
- Department of Neurobiology, Institute for Biological Research Siniša Stanković, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Aleksandra Divac Rankov
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Miguel X. Fernandes
- BioLab, Instituto Universitario de Bio-Orgánica Antonio González, Universidad de La Laguna, La Laguna, Spain
| | - Fabio Fusi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | | | - Risto Juvonen
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Magdalena Kondeva-Burdina
- Department of Pharmacology, Pharmacotherapy and Toxicology, Faculty of Pharmacy, Medical University of Sofia, Sofia, Bulgaria
| | - José M. Padrón
- BioLab, Instituto Universitario de Bio-Orgánica Antonio González, Universidad de La Laguna, La Laguna, Spain
| | - Ilza Pajeva
- Department of QSAR and Molecular Modelling, Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Tania Pencheva
- Department of QSAR and Molecular Modelling, Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Adrián Puerta
- BioLab, Instituto Universitario de Bio-Orgánica Antonio González, Universidad de La Laguna, La Laguna, Spain
| | - Hannu Raunio
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Chiara Riganti
- Department of Oncology, University of Torino, Torino, Italy
| | - Ivanka Tsakovska
- Department of QSAR and Molecular Modelling, Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Virginia Tzankova
- Department of Pharmacology, Pharmacotherapy and Toxicology, Faculty of Pharmacy, Medical University of Sofia, Sofia, Bulgaria
| | - Yordan Yordanov
- Department of Pharmacology, Pharmacotherapy and Toxicology, Faculty of Pharmacy, Medical University of Sofia, Sofia, Bulgaria
| | - Simona Saponara
- Department of Life Sciences, University of Siena, Siena, Italy
- *Correspondence: Simona Saponara,
| |
Collapse
|
91
|
Durante M, Frosini M, Chiaino E, Fusi F, Gamberucci A, Gorelli B, Chegaev K, Riganti C, Saponara S. Sdox, a H 2S releasing anthracycline, with a safer profile than doxorubicin toward vasculature. Vascul Pharmacol 2022; 143:106969. [PMID: 35149209 DOI: 10.1016/j.vph.2022.106969] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 01/24/2022] [Accepted: 02/04/2022] [Indexed: 01/18/2023]
Abstract
Sdox is a synthetic H2S-releasing doxorubicin (Dox) less cardiotoxic and more effective than Dox in pre-clinical, Dox-resistant tumour models. The well-known anthracycline vascular toxicity, however, might limit Sdox clinical use. This study aimed at evaluating Sdox vascular toxicity in vitro, using Dox as reference compound. Both vascular smooth muscle A7r5 and endothelial EA.hy926 cells were more sensitive to Dox than Sdox, although both drugs equally increased intracellular free radical levels. Sdox released H2S in both cell lines. The H2S scavenger hydroxocobalamin partially reverted Sdox-induced cytotoxicity in A7r5, but not in EA.hy926 cells, suggesting a role for H2S in smooth muscle cell death. Markers of Sdox-induced apoptosis were significantly lower than, in A7r5 cells, and comparable to those of Dox in EA.hy926 cells. In A7r5 cells, Dox increased the activity of caspase 3, 8, and 9, Sdox affecting only that of caspase 3. Moreover, both drugs induced comparable DNA damage in A7r5 cells, while Sdox was less toxic than Dox in Ea.hy926 cells. In fresh aorta rings, only Dox weakly increased phenylephrine-induced contraction when endothelium was present. In rings cultured with both drugs for 7 days, Sdox blunted phenylephrine- and high K+-induced contractions though at a concentration 10-fold higher than that of Dox. In conclusion, Sdox may represent the prototype of an innovative anthracycline, effective against Dox-resistant tumours, displaying a more favourable vascular toxicity profile compared to the parent compound.
Collapse
Affiliation(s)
- Miriam Durante
- Dipartimento di Scienze della Vita, Università di Siena, Siena, Italy
| | - Maria Frosini
- Dipartimento di Scienze della Vita, Università di Siena, Siena, Italy
| | - Elda Chiaino
- Dipartimento di Scienze della Vita, Università di Siena, Siena, Italy
| | - Fabio Fusi
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università di Siena, Siena, Italy
| | - Alessandra Gamberucci
- Dipartimento di Medicina Molecolare e dello Sviluppo, Università di Siena, Siena, Italy
| | - Beatrice Gorelli
- Dipartimento di Scienze della Vita, Università di Siena, Siena, Italy
| | - Konstantin Chegaev
- Dipartimento di Scienza e Tecnologia del Farmaco, Università di Torino, Torino, Italy
| | - Chiara Riganti
- Dipartimento di Oncologia, Università di Torino, Torino, Italy
| | - Simona Saponara
- Dipartimento di Scienze della Vita, Università di Siena, Siena, Italy.
| |
Collapse
|
92
|
Kitakata H, Endo J, Ikura H, Moriyama H, Shirakawa K, Katsumata Y, Sano M. Therapeutic Targets for DOX-Induced Cardiomyopathy: Role of Apoptosis vs. Ferroptosis. Int J Mol Sci 2022; 23:1414. [PMID: 35163335 PMCID: PMC8835899 DOI: 10.3390/ijms23031414] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/20/2022] [Accepted: 01/24/2022] [Indexed: 01/04/2023] Open
Abstract
Doxorubicin (DOX) is the most widely used anthracycline anticancer agent; however, its cardiotoxicity limits its clinical efficacy. Numerous studies have elucidated the mechanisms underlying DOX-induced cardiotoxicity, wherein apoptosis has been reported as the most common final step leading to cardiomyocyte death. However, in the past two years, the involvement of ferroptosis, a novel programmed cell death, has been proposed. The purpose of this review is to summarize the historical background that led to each form of cell death, focusing on DOX-induced cardiotoxicity and the molecular mechanisms that trigger each form of cell death. Furthermore, based on this understanding, possible therapeutic strategies to prevent DOX cardiotoxicity are outlined. DNA damage, oxidative stress, intracellular signaling, transcription factors, epigenetic regulators, autophagy, and metabolic inflammation are important factors in the molecular mechanisms of DOX-induced cardiomyocyte apoptosis. Conversely, the accumulation of lipid peroxides, iron ion accumulation, and decreased expression of glutathione and glutathione peroxidase 4 are important in ferroptosis. In both cascades, the mitochondria are an important site of DOX cardiotoxicity. The last part of this review focuses on the significance of the disruption of mitochondrial homeostasis in DOX cardiotoxicity.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Motoaki Sano
- Department of Cardiology, Keio University School of Medicine, Tokyo 160-8582, Japan; (H.K.); (J.E.); (H.I.); (H.M.); (K.S.); (Y.K.)
| |
Collapse
|
93
|
Ogunnigbagbe O, Bunick CG, Kaur K. Keratin 1 as a cell-surface receptor in cancer. Biochim Biophys Acta Rev Cancer 2022; 1877:188664. [PMID: 34890750 PMCID: PMC8818032 DOI: 10.1016/j.bbcan.2021.188664] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 11/10/2021] [Accepted: 11/30/2021] [Indexed: 01/03/2023]
Abstract
Keratins are fibrous proteins that take part in several important cellular functions, including the formation of intermediate filaments. In addition, keratins serve as epithelial cell markers, which has made their role in cancer progression, diagnosis, and treatment an important focus of research. Keratin 1 (K1) is a type II keratin whose structure is comprised of a coiled-coil central domain flanked by flexible, glycine-rich loops in the N- and C-termini. While the structure of cytoplasmic K1 is established, the structure of cell-surface K1 is not known. Several transformed cells, such as cancerous cells and cells that have undergone oxidative stress, display increased levels of overall and/or cell-surface K1 expression. Cell-surface keratins (CSKs) may be modified or truncated, and their role is yet to be fully elucidated. Current studies suggest that CSKs are involved in receptor-mediated endocytosis and immune evasion. In this Review, we discuss findings relating to K1 structure, overexpression, and cell-surface expression in the context of utilizing CSK1 as a receptor for targeted drug delivery to cancer cells, and other strategies to develop novel treatments for cancer.
Collapse
Affiliation(s)
- Oluseye Ogunnigbagbe
- School of Pharmacy, Harry and Diane Rinker Health Science Campus, Chapman University, Irvine, California, 92618-1908, USA
| | - Christopher G. Bunick
- Department of Dermatology, Yale University, New Haven, Connecticut, 06520-8059, USA,corresponding author
| | - Kamaljit Kaur
- School of Pharmacy, Harry and Diane Rinker Health Science Campus, Chapman University, Irvine, California, 92618-1908, USA,corresponding author
| |
Collapse
|
94
|
Zhang Y, Li T, Hu Y, Chen J, He Y, Gao X, Zhang Y. Co-delivery of doxorubicin and curcumin via cRGD-peptide modified PEG-PLA self-assembly nanomicelles for lung cancer therapy. CHINESE CHEM LETT 2021. [DOI: 10.1016/j.cclet.2021.11.076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
95
|
Liang Z, Chen Y, Wang Z, Wu X, Deng C, Wang C, Yang W, Tian Y, Zhang S, Lu C, Yang Y. Protective effects and mechanisms of psoralidin against adriamycin-induced cardiotoxicity. J Adv Res 2021; 40:249-261. [PMID: 36100330 PMCID: PMC9481943 DOI: 10.1016/j.jare.2021.12.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 12/12/2021] [Accepted: 12/17/2021] [Indexed: 02/08/2023] Open
|
96
|
Velasco-Ruiz A, Nuñez-Torres R, Pita G, Wildiers H, Lambrechts D, Hatse S, Delombaerde D, Van Brussel T, Alonso MR, Alvarez N, Herraez B, Vulsteke C, Zamora P, Lopez-Fernandez T, Gonzalez-Neira A. POLRMT as a Novel Susceptibility Gene for Cardiotoxicity in Epirubicin Treatment of Breast Cancer Patients. Pharmaceutics 2021; 13:1942. [PMID: 34834357 PMCID: PMC8622627 DOI: 10.3390/pharmaceutics13111942] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/06/2021] [Accepted: 11/12/2021] [Indexed: 11/27/2022] Open
Abstract
Anthracyclines are among the most used chemotherapeutic agents in breast cancer (BC). However their use is hampered by anthracycline-induced cardiotoxicity (AIC). The currently known clinical and genetic risk factors do not fully explain the observed inter-individual variability and only have a limited ability to predict which patients are more likely to develop this severe toxicity. To identify novel predictive genes, we conducted a two-stage genome-wide association study in epirubicin-treated BC patients. In the discovery phase, we genotyped over 700,000 single nucleotide variants in a cohort of 227 patients. The most interesting finding was rs62134260, located 4kb upstream of POLRMT (OR = 5.76, P = 2.23 × 10-5). We replicated this association in a validation cohort of 123 patients (P = 0.021). This variant regulates the expression of POLRMT, a gene that encodes a mitochondrial DNA-directed RNA polymerase, responsible for mitochondrial gene expression. Individuals harbouring the risk allele had a decreased expression of POLRMT in heart tissue that may cause an impaired capacity to maintain a healthy mitochondrial population in cardiomyocytes under stressful conditions, as is treatment with epirubicin. This finding suggests a novel molecular mechanism involved in the development of AIC and may improve our ability to predict patients who are at risk.
Collapse
Affiliation(s)
- Alejandro Velasco-Ruiz
- Human Genotyping Unit, CeGen (Spanish National Genotyping Centre), Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), Calle de Melchor Fernández Alamagro, 3, 28029 Madrid, Spain; (A.V.-R.); (R.N.-T.); (G.P.); (M.R.A.); (N.A.); (B.H.)
| | - Rocio Nuñez-Torres
- Human Genotyping Unit, CeGen (Spanish National Genotyping Centre), Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), Calle de Melchor Fernández Alamagro, 3, 28029 Madrid, Spain; (A.V.-R.); (R.N.-T.); (G.P.); (M.R.A.); (N.A.); (B.H.)
| | - Guillermo Pita
- Human Genotyping Unit, CeGen (Spanish National Genotyping Centre), Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), Calle de Melchor Fernández Alamagro, 3, 28029 Madrid, Spain; (A.V.-R.); (R.N.-T.); (G.P.); (M.R.A.); (N.A.); (B.H.)
| | - Hans Wildiers
- Department of General Medical Oncology, University Hospital of Leuven, Herestraat 49, 3000 Leuven, Belgium;
- Multidisciplinary Breast Centre, University Hospital of Leuven, Herestraat 49, 3000 Leuven, Belgium;
- Laboratory of Experimental Oncology (LEO), Department of Oncology, Katholieke Universiteit (KU) Leuven, Oude Markt 13, 3000 Leuven, Belgium
| | - Diether Lambrechts
- Laboratory of Translational Genetics, Centre for Cancer Biology (CCB), Flanders Institute for Biotechnology (VIB), Rijvisschestraat 120, 9052 Leuven, Belgium; (D.L.); (T.V.B.)
| | - Sigrid Hatse
- Multidisciplinary Breast Centre, University Hospital of Leuven, Herestraat 49, 3000 Leuven, Belgium;
- Laboratory of Experimental Oncology (LEO), Department of Oncology, Katholieke Universiteit (KU) Leuven, Oude Markt 13, 3000 Leuven, Belgium
| | - Danielle Delombaerde
- Integrated Cancer Center Ghent, Department of Medical Oncology, AZ Maria Middelares, 9000 Ghent, Belgium; (D.D.); (C.V.)
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, 2610 Wilrijk, Belgium
| | - Thomas Van Brussel
- Laboratory of Translational Genetics, Centre for Cancer Biology (CCB), Flanders Institute for Biotechnology (VIB), Rijvisschestraat 120, 9052 Leuven, Belgium; (D.L.); (T.V.B.)
| | - M. Rosario Alonso
- Human Genotyping Unit, CeGen (Spanish National Genotyping Centre), Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), Calle de Melchor Fernández Alamagro, 3, 28029 Madrid, Spain; (A.V.-R.); (R.N.-T.); (G.P.); (M.R.A.); (N.A.); (B.H.)
| | - Nuria Alvarez
- Human Genotyping Unit, CeGen (Spanish National Genotyping Centre), Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), Calle de Melchor Fernández Alamagro, 3, 28029 Madrid, Spain; (A.V.-R.); (R.N.-T.); (G.P.); (M.R.A.); (N.A.); (B.H.)
| | - Belen Herraez
- Human Genotyping Unit, CeGen (Spanish National Genotyping Centre), Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), Calle de Melchor Fernández Alamagro, 3, 28029 Madrid, Spain; (A.V.-R.); (R.N.-T.); (G.P.); (M.R.A.); (N.A.); (B.H.)
| | - Christof Vulsteke
- Integrated Cancer Center Ghent, Department of Medical Oncology, AZ Maria Middelares, 9000 Ghent, Belgium; (D.D.); (C.V.)
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, 2610 Wilrijk, Belgium
| | - Pilar Zamora
- Department of Medical Oncology, University Hospital La Paz, Paseo de la Castellana 261, 28046 Madrid, Spain;
| | - Teresa Lopez-Fernandez
- Department of Cardiology, University Hospital La Paz, Paseo de la Castellana 261, 28046 Madrid, Spain;
| | - Anna Gonzalez-Neira
- Human Genotyping Unit, CeGen (Spanish National Genotyping Centre), Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), Calle de Melchor Fernández Alamagro, 3, 28029 Madrid, Spain; (A.V.-R.); (R.N.-T.); (G.P.); (M.R.A.); (N.A.); (B.H.)
| |
Collapse
|
97
|
Qiu S, Zhou T, Qiu B, Zhang Y, Zhou Y, Yu H, Zhang J, Liu L, Yuan L, Yang G, Duan Y, Xing C. Risk Factors for Anthracycline-Induced Cardiotoxicity. Front Cardiovasc Med 2021; 8:736854. [PMID: 34660739 PMCID: PMC8511483 DOI: 10.3389/fcvm.2021.736854] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 08/30/2021] [Indexed: 11/30/2022] Open
Abstract
Background: Several cardiovascular risk factors have been suggested to be associated with anthracycline-induced cardiotoxicity, but their quantitative effects have not reached a consensus. Methods: We searched PubMed, EMBASE, and Cochrane Library databases for manuscripts published from inception to February 2021, which reported the results of cardiotoxicity due to anthracycline chemotherapy without trastuzumab. Cardiotoxicity defined by any reduction of left ventricular eject fraction (LVEF) to below 50% or a >10% reduction from baseline was defined as the primary endpoint. Odd ratios (OR) with 95% confidence intervals (CI) were calculated using a random-effects model meta-analysis. Results: A total of 7,488 patients receiving anthracycline chemotherapy without trastuzumab were included, who had at least one risk factor at baseline. Hypertension (OR: 1.99; 95% CI: 1.43–2.76), diabetes mellitus (OR: 1.74; 95% CI: 1.11–2.74), and obesity (OR: 1.72; 95% CI: 1.13–2.61) were associated with increased risk of cardiotoxicity. In addition, the relative reduction of global longitudinal strain (GLS) from baseline after anthracycline treatment could significantly improve the detection ability of cardiotoxicity (28.5%, 95% CI: 22.1–35.8% vs. 16.4%, 95% CI: 13.4–19.9%) compared with LVEF. The early detection rate of anthracycline-induced cardiotoxicity (3 months after chemotherapy) by GLS was 30.2% (95% CI: 24.9–36.1%), which is similar with the overall result of GLS. Conclusions: Hypertension, diabetes mellitus, and obesity are associated with increased risk of anthracycline-induced cardiotoxicity, which indicates that corresponding protective strategies should be used during and after anthracycline treatment. The findings of higher detection rate and better early detection ability for cardiotoxicity than LVEF added new proofs for the advantages of GLS in detection of AIC.
Collapse
Affiliation(s)
- Shuo Qiu
- Department of Ultrasound Diagnostics, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Tian Zhou
- Department of Ultrasound Diagnostics, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Bo Qiu
- Department of General Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Yuxin Zhang
- Department of Ultrasound Diagnostics, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Yonggang Zhou
- Department of Ultrasound Diagnostics, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Huihui Yu
- School of Nursing, Air Force Medical University, Xi'an, China
| | - Jingyi Zhang
- Department of Hematology, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Li Liu
- Department of Hematology, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Lijun Yuan
- Department of Ultrasound Diagnostics, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Guodong Yang
- Department of Biochemistry and Molecular Biology, Air Force Medical University, Xi'an, China
| | - Yunyou Duan
- Department of Ultrasound Diagnostics, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Changyang Xing
- Department of Ultrasound Diagnostics, Tangdu Hospital, Air Force Medical University, Xi'an, China
| |
Collapse
|
98
|
Costa M, Blaschke TF, Amara SG, Meyer UA, Insel PA. Introduction to the Theme "Old and New Toxicology: Interfaces with Pharmacology". Annu Rev Pharmacol Toxicol 2021; 61:1-7. [PMID: 33411582 DOI: 10.1146/annurev-pharmtox-092220-033032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The theme of Volume 61 is "Old and New Toxicology: Interfaces with Pharmacology." Old toxicology is exemplified by the authors of the autobiographical articles: B.M. Olivera's work on toxins and venoms from cone snails and P. Taylor's studies of acetylcholinesterase and the nicotinic cholinergic receptor, which serve as sites of action for numerous pesticides and venoms. Other articles in this volume focus on new understanding and new types of toxicology, including (a) arsenic toxicity, which is an ancient poison that, through evolution, has caused most multicellular organisms to express an active arsenic methyltransferase to methylate arsenite, which accelerates the excretion of arsenic from the body; (b) small molecules that react with lipid dicarbonyls, which are now considered the most toxic oxidative stress end products; (c) immune checkpoint inhibitors (ICIs), which have revolutionized cancer therapy but have numerous immune-related adverse events, including cardiovascular complications; (d) autoimmunity caused by the environment; (e) idiosyncratic drug-induced liver disease, which together with the toxicity of ICIs represents new toxicology interfacing with pharmacology; and (f) sex differences in the development of cardiovascular disease, with men more susceptible than women to vascular inflammation that initiates and perpetuates disease. These articles and others in Volume 61 reflect the interface and close integration of pharmacology and toxicology that began long ago but continues today.
Collapse
Affiliation(s)
- Max Costa
- Department of Environmental Medicine, NYU Grossman School of Medicine, New York, New York 10010, USA;
| | | | - Susan G Amara
- National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Urs A Meyer
- Biozentrum, University of Basel, CH-4056 Basel, Switzerland
| | - Paul A Insel
- Departments of Pharmacology and Medicine, University of California, San Diego, La Jolla, California 92093, USA
| |
Collapse
|
99
|
Li Y, Yang Y, Shao Y, Sun Y, Si H, Miao J, Xu Y. Chitosan functionalized graphene oxide nanocomposites for fluorescence imaging of apoptotic processes and targeted anti-inflammation study. Carbohydr Polym 2021; 269:118345. [PMID: 34294352 DOI: 10.1016/j.carbpol.2021.118345] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 06/01/2021] [Accepted: 06/11/2021] [Indexed: 12/25/2022]
Abstract
This work reports novel chitosan functionalized graphene oxide (GO) nanocomposites combined fluorescence imaging and therapeutic functions in one agent, which can serve as a promising alternative to alleviate related diseases caused hyperinflammation. Briefly, GO was designed to be conjugated with chitosan, fluorescein-labeled peptide, toll-like receptor 4 antibody and hydroxycamptothecin/aloe emodin. We have demonstrated that such nanocomposites could effectively achieve active targeted delivery of pro-apoptotic and anti-inflammatory drugs into inflammatory cells and cause cells apoptosis by acid-responsive drug release. Moreover, confocal fluorescence imaging confirms that the drug-induced inflammatory cells apoptosis could be visualized the light-up fluorescence of fluorescein activated by caspase-3. Meanwhile, inflammatory-related biomarkers have down-regulated after the nanocomposites' treatment in both vitro and vivo experiments consistent with the results in histological sections. In summary, the bifunctional nanocomposites that possess anti-inflammation and fluorescence imaging could serve as a promising therapeutic agent for reducing hyperinflammation caused by numerous diseases.
Collapse
Affiliation(s)
- Yi Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Yazhi Yang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Yingge Shao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Yangyang Sun
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Hongbin Si
- College of Animal Sciences and Technology, Guangxi University, Nanning, Guangxi 530004, China
| | - Jinfeng Miao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Yuanyuan Xu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
100
|
Russo M, Bono E, Ghigo A. The Interplay Between Autophagy and Senescence in Anthracycline Cardiotoxicity. Curr Heart Fail Rep 2021; 18:180-190. [PMID: 34081265 PMCID: PMC8342382 DOI: 10.1007/s11897-021-00519-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/19/2021] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Doxorubicin (DOXO) is a highly effective chemotherapeutic drug employed for the treatment of a wide spectrum of cancers, spanning from solid tumours to haematopoietic malignancies. However, its clinical use is hampered by severe and dose-dependent cardiac side effects that ultimately lead to heart failure (HF). RECENT FINDINGS Mitochondrial dysfunction and oxidative stress are well-established mechanisms of DOXO-induced cardiotoxicity, although recent evidence suggests that deregulation of other biological processes, like autophagy, could be involved. It is increasingly recognized that autophagy deregulation is intimately interconnected with the initiation of detrimental cellular responses, including autosis and senescence, raising the possibility of using autophagy modulators as well as senolytics and senomorphics for preventing DOXO cardiotoxicity. This review aims at providing an overview of the signalling pathways that are common to autophagy and senescence, with a special focus on how the relationship between these two processes is deregulated in response to cardiotoxic treatments. Finally, we will discuss the potential therapeutic utility of drugs modulating autophagy and/or senescence for counteracting DOXO cardiotoxicity.
Collapse
Affiliation(s)
- Michele Russo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Enrico Bono
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126, Torino, Italy.
| |
Collapse
|